1
|
Pawluk H, Tafelska-Kaczmarek A, Sopońska M, Porzych M, Modrzejewska M, Pawluk M, Kurhaluk N, Tkaczenko H, Kołodziejska R. The Influence of Oxidative Stress Markers in Patients with Ischemic Stroke. Biomolecules 2024; 14:1130. [PMID: 39334896 PMCID: PMC11430825 DOI: 10.3390/biom14091130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/27/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024] Open
Abstract
Stroke is the second leading cause of death worldwide, and its incidence is rising rapidly. Acute ischemic stroke is a subtype of stroke that accounts for the majority of stroke cases and has a high mortality rate. An effective treatment for stroke is to minimize damage to the brain's neural tissue by restoring blood flow to decreased perfusion areas of the brain. Many reports have concluded that both oxidative stress and excitotoxicity are the main pathological processes associated with ischemic stroke. Current measures to protect the brain against serious damage caused by stroke are insufficient. For this reason, it is important to investigate oxidative and antioxidant strategies to reduce oxidative damage. This review focuses on studies assessing the concentration of oxidative stress biomarkers and the level of antioxidants (enzymatic and non-enzymatic) and their impact on the clinical prognosis of patients after stroke. Mechanisms related to the production of ROS/RNS and the role of oxidative stress in the pathogenesis of ischemic stroke are presented, as well as new therapeutic strategies aimed at reducing the effects of ischemia and reperfusion.
Collapse
Affiliation(s)
- Hanna Pawluk
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Karlowicza 24, 85-092 Bydgoszcz, Poland
| | - Agnieszka Tafelska-Kaczmarek
- Department of Organic Chemistry, Faculty of Chemistry, Nicolaus Copernicus University, Gagarina 7, 87-100 Torun, Poland
| | - Małgorzata Sopońska
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Karlowicza 24, 85-092 Bydgoszcz, Poland
| | - Marta Porzych
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Karlowicza 24, 85-092 Bydgoszcz, Poland
| | - Martyna Modrzejewska
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Karlowicza 24, 85-092 Bydgoszcz, Poland
| | - Mateusz Pawluk
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Karlowicza 24, 85-092 Bydgoszcz, Poland
| | - Natalia Kurhaluk
- Institute of Biology, Pomeranian University in Slupsk, Arciszewski 22B, 76-200 Slupsk, Poland
| | - Halina Tkaczenko
- Institute of Biology, Pomeranian University in Slupsk, Arciszewski 22B, 76-200 Slupsk, Poland
| | - Renata Kołodziejska
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Karlowicza 24, 85-092 Bydgoszcz, Poland
| |
Collapse
|
2
|
Fan G, Liu M, Liu J, Huang Y, Mu W. Traditional Chinese medicines treat ischemic stroke and their main bioactive constituents and mechanisms. Phytother Res 2024; 38:411-453. [PMID: 38051175 DOI: 10.1002/ptr.8033] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 09/12/2023] [Accepted: 09/24/2023] [Indexed: 12/07/2023]
Abstract
Ischemic stroke (IS) remains one of the leading causes of death and disability in humans. Unfortunately, none of the treatments effectively provide functional benefits to patients with IS, although many do so by targeting different aspects of the ischemic cascade response. The advantages of traditional Chinese medicine (TCM) in preventing and treating IS are obvious in terms of early treatment and global coordination. The efficacy of TCM and its bioactive constituents has been scientifically proven over the past decades. Based on clinical trials, this article provides a review of commonly used TCM patent medicines and herbal decoctions indicated for IS. In addition, this paper also reviews the mechanisms of bioactive constituents in TCM for the treatment of IS in recent years, both domestically and internationally. A comprehensive review of preclinical and clinical studies will hopefully provide new ideas to address the threat of IS.
Collapse
Affiliation(s)
- Genhao Fan
- Tianjin University of Chinese Medicine, Tianjin, China
- Clinical Pharmacology Department, Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Menglin Liu
- Tianjin University of Chinese Medicine, Tianjin, China
| | - Jia Liu
- Clinical Pharmacology Department, Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuhong Huang
- Clinical Pharmacology Department, Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wei Mu
- Clinical Pharmacology Department, Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
3
|
Zhou M, Xin J, Chen J, Sun C, Huo B, Zhang W, Liu X. Scientific Landscape of Oxidative Stress in Stroke: From a Bibliometric Analysis to an in-Depth Review. Neurochem Res 2023; 48:3327-3348. [PMID: 37505366 DOI: 10.1007/s11064-023-03999-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 07/16/2023] [Accepted: 07/18/2023] [Indexed: 07/29/2023]
Abstract
Stroke is an acute cerebrovascular disease resulting from either obstruction or rupture of a blood vessel in the brain. Oxidative stress (OS), referred to a status where cellular oxidative capacities overwhelm antioxidative defenses, is involved in the pathophysiology of stroke. The bibliometric analysis and in-depth review aim to depict the research trend of OS in stroke. Relevant scientific publications were acquired from the Web of Science Core Collection database. Scientific landscape of OS in stroke was illustrated by general quantitative trend, impactful journals, and co-authorship of various academic units (i.e., countries/regions, organizations, and authors). Furthermore, theme analysis predicting the hot research issues and frontiers was performed. 15,826 documents regarding OS in stroke were obtained over a time span of more than 20 years from 1992 to 2021. The overall tendency of publication counts was continuously on the rise. Bibliometric analysis indicated China and the United States were predominant in this study field, as reflected by their high publication counts and intensive collaboration with other countries. Current key research areas of OS in stroke may lie in the investigation of neuroinflammation, and interaction among multiple cell death mechanisms including apoptosis, autophagy, and ferroptosis to search for effective treatments. Moreover, another hot topic could be the association between air pollution and stroke, and its underlying mechanisms. As the exploration of OS in stroke is speculated to be a continuous hot spot in the future, this article may be helpful for researchers to conduct future studies with the understanding of influential academic forces and research highlights.
Collapse
Affiliation(s)
- Minqi Zhou
- Department of Histology and Embryology, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hang Kong Road, Wuhan, 430030, China
| | - Jiayu Xin
- Department of Histology and Embryology, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hang Kong Road, Wuhan, 430030, China
| | - Jinyu Chen
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Caiyun Sun
- Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan, 430081, China
| | - Bingyue Huo
- Department of Histology and Embryology, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hang Kong Road, Wuhan, 430030, China
| | - Wenting Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hang Kong Road, Wuhan, 430030, China
| | - Xiangqian Liu
- Department of Histology and Embryology, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hang Kong Road, Wuhan, 430030, China.
| |
Collapse
|
4
|
You Q, Lan XB, Liu N, Du J, Ma L, Yang JM, Niu JG, Peng XD, Jin GL, Yu JQ. Neuroprotective strategies for neonatal hypoxic-ischemic brain damage: Current status and challenges. Eur J Pharmacol 2023; 957:176003. [PMID: 37640219 DOI: 10.1016/j.ejphar.2023.176003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/17/2023] [Accepted: 08/18/2023] [Indexed: 08/31/2023]
Abstract
Neonatal hypoxic-ischemic brain damage (HIBD) is a prominent contributor to both immediate mortality and long-term impairment in newborns. The elusive nature of the underlying mechanisms responsible for neonatal HIBD presents a significant obstacle in the effective clinical application of numerous pharmaceutical interventions. This comprehensive review aims to concentrate on the potential neuroprotective agents that have demonstrated efficacy in addressing various pathogenic factors associated with neonatal HIBD, encompassing oxidative stress, calcium overload, mitochondrial dysfunction, endoplasmic reticulum stress, inflammatory response, and apoptosis. In this review, we conducted an analysis of the precise molecular pathways by which these drugs elicit neuroprotective effects in animal models of neonatal hypoxic-ischemic brain injury (HIBD). Our objective was to provide a comprehensive overview of potential neuroprotective agents for the treatment of neonatal HIBD in animal experiments, with the ultimate goal of enhancing the feasibility of clinical translation and establishing a solid theoretical foundation for the clinical management of neonatal HIBD.
Collapse
Affiliation(s)
- Qing You
- Department of Pharmacology, School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China.
| | - Xiao-Bing Lan
- Department of Pharmacology, School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China.
| | - Ning Liu
- Department of Pharmacology, School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China; Ningxia Special Traditional Medicine Modern Engineering Research Center and Collaborative Innovation Center, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China.
| | - Juan Du
- Department of Pharmacology, School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China.
| | - Lin Ma
- Department of Pharmacology, School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China.
| | - Jia-Mei Yang
- Department of Pharmacology, School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China.
| | - Jian-Guo Niu
- Ningxia Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, 750004, China.
| | - Xiao-Dong Peng
- Department of Pharmacology, School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China.
| | - Gui-Lin Jin
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fuzhou, 350108, Fujian, China; Department of Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, 350108, Fujian, China.
| | - Jian-Qiang Yu
- Department of Pharmacology, School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China; Ningxia Special Traditional Medicine Modern Engineering Research Center and Collaborative Innovation Center, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China.
| |
Collapse
|
5
|
Kaiser EE, Waters ES, Yang X, Fagan MM, Scheulin KM, Sneed SE, Cheek SR, Jeon JH, Shin SK, Kinder HA, Kumar A, Platt SR, Duberstein KJ, Park HJ, Xie J, West FD. Tanshinone IIA-Loaded Nanoparticle and Neural Stem Cell Therapy Enhances Recovery in a Pig Ischemic Stroke Model. Stem Cells Transl Med 2022; 11:1061-1071. [PMID: 36124817 PMCID: PMC9585947 DOI: 10.1093/stcltm/szac062] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 07/17/2022] [Indexed: 12/30/2022] Open
Abstract
Induced pluripotent stem cell-derived neural stem cells (iNSCs) are a multimodal stroke therapeutic that possess neuroprotective, regenerative, and cell replacement capabilities post-ischemia. However, long-term engraftment and efficacy of iNSCs is limited by the cytotoxic microenvironment post-stroke. Tanshinone IIA (Tan IIA) is a therapeutic that demonstrates anti-inflammatory and antioxidative effects in rodent ischemic stroke models and stroke patients. Therefore, pretreatment with Tan IIA may create a microenvironment that is more conducive to the long-term survival of iNSCs. In this study, we evaluated the potential of Tan IIA drug-loaded nanoparticles (Tan IIA-NPs) to improve iNSC engraftment and efficacy, thus potentially leading to enhanced cellular, tissue, and functional recovery in a translational pig ischemic stroke model. Twenty-two pigs underwent middle cerebral artery occlusion (MCAO) and were randomly assigned to a PBS + PBS, PBS + iNSC, or Tan IIA-NP + iNSC treatment group. Magnetic resonance imaging (MRI), modified Rankin Scale neurological evaluation, and immunohistochemistry were performed over a 12-week study period. Immunohistochemistry indicated pretreatment with Tan IIA-NPs increased iNSC survivability. Furthermore, Tan IIA-NPs increased iNSC neuronal differentiation and decreased iNSC reactive astrocyte differentiation. Tan IIA-NP + iNSC treatment enhanced endogenous neuroprotective and regenerative activities by decreasing the intracerebral cellular immune response, preserving endogenous neurons, and increasing neuroblast formation. MRI assessments revealed Tan IIA-NP + iNSC treatment reduced lesion volumes and midline shift. Tissue preservation and recovery corresponded with significant improvements in neurological recovery. This study demonstrated pretreatment with Tan IIA-NPs increased iNSC engraftment, enhanced cellular and tissue recovery, and improved neurological function in a translational pig stroke model.
Collapse
Affiliation(s)
- Erin E Kaiser
- Regenerative Bioscience Center, Athens, GA, USA
- Biomedical and Health Sciences Institute, Athens, GA, USA
- Animal and Dairy Science Department, College of Agricultural and Environmental Sciences, Athens, GA, USA
| | - Elizabeth S Waters
- Regenerative Bioscience Center, Athens, GA, USA
- Biomedical and Health Sciences Institute, Athens, GA, USA
- Animal and Dairy Science Department, College of Agricultural and Environmental Sciences, Athens, GA, USA
- Environmental Health Science Department, College of Public Health, Athens, GA, USA
| | - Xueyuan Yang
- Chemistry Department, Franklin College of Arts and Sciences, Athens, GA, USA
| | - Madison M Fagan
- Regenerative Bioscience Center, Athens, GA, USA
- Biomedical and Health Sciences Institute, Athens, GA, USA
- Animal and Dairy Science Department, College of Agricultural and Environmental Sciences, Athens, GA, USA
| | - Kelly M Scheulin
- Regenerative Bioscience Center, Athens, GA, USA
- Biomedical and Health Sciences Institute, Athens, GA, USA
- Animal and Dairy Science Department, College of Agricultural and Environmental Sciences, Athens, GA, USA
| | - Sydney E Sneed
- Regenerative Bioscience Center, Athens, GA, USA
- Animal and Dairy Science Department, College of Agricultural and Environmental Sciences, Athens, GA, USA
| | | | - Julie Heejin Jeon
- Nutritional Sciences Department, College of Family and Consumer Sciences, Athens, GA, USA
| | - Soo K Shin
- Regenerative Bioscience Center, Athens, GA, USA
- Animal and Dairy Science Department, College of Agricultural and Environmental Sciences, Athens, GA, USA
- Small Animal Medicine and Surgery Department, College of Veterinary Medicine, Athens, GA, USA
| | - Holly A Kinder
- Regenerative Bioscience Center, Athens, GA, USA
- Biomedical and Health Sciences Institute, Athens, GA, USA
- Animal and Dairy Science Department, College of Agricultural and Environmental Sciences, Athens, GA, USA
| | - Anil Kumar
- Chemistry Department, Franklin College of Arts and Sciences, Athens, GA, USA
| | - Simon R Platt
- Regenerative Bioscience Center, Athens, GA, USA
- Interdisciplinary Toxicology Program, College of Pharmacy, University of Georgia, Athens, GA, USA
| | - Kylee J Duberstein
- Regenerative Bioscience Center, Athens, GA, USA
- Animal and Dairy Science Department, College of Agricultural and Environmental Sciences, Athens, GA, USA
| | - Hea Jin Park
- Nutritional Sciences Department, College of Family and Consumer Sciences, Athens, GA, USA
| | - Jin Xie
- Regenerative Bioscience Center, Athens, GA, USA
- Chemistry Department, Franklin College of Arts and Sciences, Athens, GA, USA
| | - Franklin D West
- Regenerative Bioscience Center, Athens, GA, USA
- Biomedical and Health Sciences Institute, Athens, GA, USA
- Animal and Dairy Science Department, College of Agricultural and Environmental Sciences, Athens, GA, USA
- Small Animal Medicine and Surgery Department, College of Veterinary Medicine, Athens, GA, USA
| |
Collapse
|
6
|
Huang X, Jin L, Deng H, Wu D, Shen QK, Quan ZS, Zhang CH, Guo HY. Research and Development of Natural Product Tanshinone I: Pharmacology, Total Synthesis, and Structure Modifications. Front Pharmacol 2022; 13:920411. [PMID: 35903340 PMCID: PMC9315943 DOI: 10.3389/fphar.2022.920411] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/08/2022] [Indexed: 11/13/2022] Open
Abstract
Salvia miltiorrhiza (S. miltiorrhiza), which has been used for thousands of years to treat cardiovascular diseases, is a well-known Chinese medicinal plant. The fat-soluble tanshinones in S. miltiorrhiza are important biologically active ingredients including tanshinone I, tanshinone IIA, dihydrotanshinone, and cryptotanshinone. Tanshinone I, a natural diterpenoid quinone compound widely used in traditional Chinese medicine, has a wide range of biological effects including anti-cancer, antioxidant, neuroprotective, and anti-inflammatory activities. To further improve its potency, water solubility, and bioavailability, tanshinone I can be used as a platform for drug discovery to generate high-quality drug candidates with unique targets and enhanced drug properties. Numerous derivatives of tanshinone I have been developed and have contributed to major advances in the identification of new drugs to treat human cancers and other diseases and in the study of related molecular mechanisms. This review focuses on the structural modification, total synthesis, and pharmacology of tanshinone I. We hope that this review will help understanding the research progress in this field and provide constructive suggestions for further research on tanshinone I.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Hong-Yan Guo
- *Correspondence: Chang-hao Zhang, ; Hong-Yan Guo,
| |
Collapse
|
7
|
Sheida A, Taghavi T, Shafabakhsh R, Ostadian A, Razaghi Bahabadi Z, Khaksary Mahabady M, Hamblin MR, Mirzaei H. Potential of natural products in the treatment of myocardial infarction: focus on molecular mechanisms. Crit Rev Food Sci Nutr 2022; 63:5488-5505. [PMID: 34978223 DOI: 10.1080/10408398.2021.2020720] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Although conventional drugs are widely used in the prevention and treatment of cardiovascular disease (CVD), they are being used less frequently due to concerns about possible side effects over the long term. There has been a renewed research interest in medicinal plant products, and their role in protecting the cardiovascular system and treating CVD, which are now being considered as potential alternatives to modern drugs. The most important mechanism causing damage to the myocardium after heart attack and reperfusion, is increased levels of free radicals and oxidative stress. Therefore, treatment approaches often focus on reducing free radicals or enhancing antioxidant defense mechanism. It has been previously reported that bioactive natural products can protect the heart muscle in myocardial infarction (MI). Since these compounds are readily available in fruits and vegetables, they could prevent the risk of MI if they are consumed daily. Although the benefits of a healthy diet are well known, many scientific studies have focused on whether pure natural compounds can prevent and treat MI. In this review we summarize the effects of curcumin, resveratrol, quercitin, berberine, and tanshinone on MI and CVD, and focus on their proposed molecular mechanisms of action.
Collapse
Affiliation(s)
- Amirhossein Sheida
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Rana Shafabakhsh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Amirreza Ostadian
- Department of Laboratory Medicine, School of Allied Medical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Zahra Razaghi Bahabadi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Mahmood Khaksary Mahabady
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, South Africa
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
8
|
OUP accepted manuscript. J Pharm Pharmacol 2022; 74:1230-1240. [DOI: 10.1093/jpp/rgac012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 02/12/2022] [Indexed: 11/14/2022]
|
9
|
Therapeutic Effects of Risperidone against Spinal Cord Injury in a Rat Model of Asphyxial Cardiac Arrest: A Focus on Body Temperature, Paraplegia, Motor Neuron Damage, and Neuroinflammation. Vet Sci 2021; 8:vetsci8100230. [PMID: 34679060 PMCID: PMC8537088 DOI: 10.3390/vetsci8100230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/06/2021] [Accepted: 10/08/2021] [Indexed: 11/17/2022] Open
Abstract
Cardiac arrest (CA) causes severe spinal cord injury and evokes spinal cord disorders including paraplegia. It has been reported that risperidone, an antipsychotic drug, effectively protects neuronal cell death from transient ischemia injury in gerbil brains. However, until now, studies on the effects of risperidone on spinal cord injury after asphyxial CA (ACA) and cardiopulmonary resuscitation (CPR) are not sufficient. Therefore, this study investigated the effect of risperidone on hind limb motor deficits and neuronal damage/death in the lumbar part of the spinal cord following ACA in rats. Mortality, severe motor deficits in the hind limbs, and the damage/death (loss) of motor neurons located in the anterior horn were observed two days after ACA/CPR. These symptoms were significantly alleviated by risperidone (an atypical antipsychotic) treatment after ACA. In vehicle-treated rats, the immunoreactivities of tumor necrosis factor-alpha (TNF-α) and interleukin 1-beta (IL-1β), as pro-inflammatory cytokines, were increased, and the immunoreactivities of IL-4 and IL-13, as anti-inflammatory cytokines, were reduced with time after ACA/CPR. In contrast, in risperidone-treated rats, the immunoreactivity of the pro-inflammatory cytokines was significantly decreased, and the anti-inflammatory cytokines were enhanced compared to vehicle-treated rats. In brief, risperidone treatment after ACA/CPR in rats significantly improved the survival rate and attenuated paralysis, the damage/death (loss) of motor neurons, and inflammation in the lumbar anterior horn. Thus, risperidone might be a therapeutic agent for paraplegia by attenuation of the damage/death (loss) of spinal motor neurons and neuroinflammation after ACA/CPR.
Collapse
|
10
|
Prajapati R, Park SE, Seong SH, Paudel P, Fauzi FM, Jung HA, Choi JS. Monoamine Oxidase Inhibition by Major Tanshinones from Salvia miltiorrhiza and Selective Muscarinic Acetylcholine M 4 Receptor Antagonism by Tanshinone I. Biomolecules 2021; 11:1001. [PMID: 34356625 PMCID: PMC8301926 DOI: 10.3390/biom11071001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/30/2021] [Accepted: 07/05/2021] [Indexed: 11/23/2022] Open
Abstract
Monoamine oxidases (MAOs) and muscarinic acetylcholine receptors (mAChRs) are considered important therapeutic targets for Parkinson's disease (PD). Lipophilic tanshinones are major phytoconstituents in the dried roots of Salvia miltiorrhiza that have demonstrated neuroprotective effects against dopaminergic neurotoxins and the inhibition of MAO-A. Since MAO-B inhibition is considered an effective therapeutic strategy for PD, we tested the inhibitory activities of three abundant tanshinone congeners against recombinant human MAO (hMAO) isoenzymes through in vitro experiments. In our study, tanshinone I (1) exhibited the highest potency against hMAO-A, followed by tanshinone IIA and cryptotanshinone, with an IC50 less than 10 µM. They also suppressed hMAO-B activity, with an IC50 below 25 µM. Although tanshinones are known to inhibit hMAO-A, their enzyme inhibition mechanism and binding sites have yet to be investigated. Enzyme kinetics and molecular docking studies have revealed the mode of inhibition and interactions of tanshinones during enzyme inhibition. Proteochemometric modeling predicted mAChRs as possible pharmacological targets of 1, and in vitro functional assays confirmed the selective M4 antagonist nature of 1 (56.1% ± 2.40% inhibition of control agonist response at 100 µM). These findings indicate that 1 is a potential therapeutic molecule for managing the motor dysfunction and depression associated with PD.
Collapse
Affiliation(s)
- Ritu Prajapati
- Department of Food and Life Science, Pukyong National University, Busan 48513, Korea; (R.P.); (S.E.P.); (S.H.S.); (P.P.)
| | - Se Eun Park
- Department of Food and Life Science, Pukyong National University, Busan 48513, Korea; (R.P.); (S.E.P.); (S.H.S.); (P.P.)
- Department of Biomedical Science, Asan Medical Institute of Convergence Science and Technology, University of Ulsan, Seoul 05505, Korea
| | - Su Hui Seong
- Department of Food and Life Science, Pukyong National University, Busan 48513, Korea; (R.P.); (S.E.P.); (S.H.S.); (P.P.)
- Natural Product Research Division, Honam National Institute of Biological Resource, Mokpo 58762, Korea
| | - Pradeep Paudel
- Department of Food and Life Science, Pukyong National University, Busan 48513, Korea; (R.P.); (S.E.P.); (S.H.S.); (P.P.)
- National Center for Natural Products Research, Research Institute of Pharmaceutical Science, The University of Mississippi, Oxford, MS 38677, USA
| | - Fazlin Mohd Fauzi
- Department of Pharmacology and Chemistry, Faculty of Pharmacy, Universiti Teknologi MARA, Puncak Alam 42300, Malaysia;
| | - Hyun Ah Jung
- Department of Food Science and Human Nutrition, Jeonbok National University, Jeonju 54896, Korea
| | - Jae Sue Choi
- Department of Food and Life Science, Pukyong National University, Busan 48513, Korea; (R.P.); (S.E.P.); (S.H.S.); (P.P.)
| |
Collapse
|
11
|
Baicalein, Baicalin, and Wogonin: Protective Effects against Ischemia-Induced Neurodegeneration in the Brain and Retina. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8377362. [PMID: 34306315 PMCID: PMC8263226 DOI: 10.1155/2021/8377362] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/08/2021] [Accepted: 06/19/2021] [Indexed: 12/17/2022]
Abstract
Ischemia is a common pathological condition present in many neurodegenerative diseases, including ischemic stroke, retinal vascular occlusion, diabetic retinopathy, and glaucoma, threatening the sight and lives of millions of people globally. Ischemia can trigger excessive oxidative stress, inflammation, and vascular dysfunction, leading to the disruption of tissue homeostasis and, ultimately, cell death. Current therapies are very limited and have a narrow time window for effective treatment. Thus, there is an urgent need to develop more effective therapeutic options for ischemia-induced neural injuries. With emerging reports on the pharmacological properties of natural flavonoids, these compounds present potent antioxidative, anti-inflammatory, and antiapoptotic agents for the treatment of ischemic insults. Three major active flavonoids, baicalein, baicalin, and wogonin, have been extracted from Scutellaria baicalensis Georgi (S. baicalensis); all of which are reported to have low cytotoxicity. They have been demonstrated to exert promising pharmacological capabilities in preventing cell and tissue damage. This review focuses on the therapeutic potentials of these flavonoids against ischemia-induced neurotoxicity and damage in the brain and retina. The bioactivity and bioavailability of baicalein, baicalin, and wogonin are also discussed. It is with hope that the therapeutic potential of these flavonoids can be utilized and developed as natural treatments for ischemia-induced injuries of the central nervous system (CNS).
Collapse
|
12
|
Liu J, Wang F, Sheng P, Xia Z, Jiang Y, Yan BC. A network-based method for mechanistic investigation and neuroprotective effect on treatment of tanshinone Ⅰ against ischemic stroke in mouse. JOURNAL OF ETHNOPHARMACOLOGY 2021; 272:113923. [PMID: 33617968 DOI: 10.1016/j.jep.2021.113923] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/22/2021] [Accepted: 02/09/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tanshinone-Ⅰ (TSNⅠ), a member of the mainly active components of Salvia miltiorrhiza Bunge (Dan Shen), which is widely used for the treatment for modern clinical diseases including cardiovascular and cerebrovascular diseases, has been reported to show the properties of anti-oxidation, anti-inflammation, neuroprotection and other pharmacological actions. However, whether TSNⅠ can improve neuron survival and neurological function against transient focal cerebral ischemia (tMCAO) in mice is still a blank field. AIM OF THE STUDY This study aims to investigate the neuroprotective effects of TSNⅠ on ischemic stroke (IS) induced by tMCAO in mice and explore the potential mechanism of TSNⅠ against IS by combining network pharmacology approach and experimental verification. MATERIALS AND METHODS In this study, the pivotal candidate targets of TSNⅠ against IS were screened by network pharmacology firstly. Enrichment analysis and molecular docking of those targets were performed to identify the possible mechanism of TSNⅠ against IS. Afterwards, experiments were carried out to further verify the mechanism of TSNⅠ against IS. The infarct volume and neurological deficit were evaluated by 2, 3, 5-triphenyl tetrazolium chloride (TTC) staining and Longa respectively. Immunohistochemistry was used to observe neuronal death in the hippocampus and cortical regions by detecting the change of NeuN. The predicting pathways of signaling-related proteins were assessed by Western blot in vitro and in vivo experiments. RESULTS In vivo, TSNⅠ was found to dose-dependently decrease mice's cerebral infarct volume induced by tMCAO. In vitro, pretreatment with TSNⅠ could increase cell viability of HT-22 cell following oxygen-glucose deprivation (OGD/R). Moreover, the results showed that 125 candidate targets were identified, Protein kinase B (AKT) signaling pathway was significantly enriched by Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis and mitogen-activated protein kinases 1 (MAPK1) and AKT1 could be bound to TSNⅠ more firmly by molecular docking analysis, which implies that TSNⅠ may play a role in neuroprotection through activating AKT and MAPK signaling pathways. Meanwhile, TSNⅠ was confirmed to significantly protect neurons from injury induced by IS through activating AKT and MAPK signaling pathways. CONCLUSION In conclusion, our study clarifies that the mechanism of TSNⅠ against IS might be related to AKT and MAPK signaling pathways, which may provide the basic evidence for further development and utilization of TSNⅠ.
Collapse
Affiliation(s)
- Jiajia Liu
- Medical College, Institute of Translational Medicine, Department of Neurology, Affiliated Hospital of Yangzhou University, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou University, Yangzhou, 225001, PR China
| | - Fuxing Wang
- Medical College, Institute of Translational Medicine, Department of Neurology, Affiliated Hospital of Yangzhou University, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou University, Yangzhou, 225001, PR China
| | - Peng Sheng
- Medical College, Institute of Translational Medicine, Department of Neurology, Affiliated Hospital of Yangzhou University, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou University, Yangzhou, 225001, PR China
| | - Zihao Xia
- Medical College, Institute of Translational Medicine, Department of Neurology, Affiliated Hospital of Yangzhou University, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou University, Yangzhou, 225001, PR China
| | - Yunyao Jiang
- School of Pharmaceutical Sciences, Institute for Chinese Materia Medica, Tsinghua University, Beijing, 100084, PR China
| | - Bing Chun Yan
- Medical College, Institute of Translational Medicine, Department of Neurology, Affiliated Hospital of Yangzhou University, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou University, Yangzhou, 225001, PR China; Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, PR China.
| |
Collapse
|
13
|
Subedi L, Gaire BP. Tanshinone IIA: A phytochemical as a promising drug candidate for neurodegenerative diseases. Pharmacol Res 2021; 169:105661. [PMID: 33971269 DOI: 10.1016/j.phrs.2021.105661] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 04/02/2021] [Accepted: 04/30/2021] [Indexed: 02/07/2023]
Abstract
Tanshinones, lipophilic diterpenes isolated from the rhizome of Salvia miltiorrhiza, have diverse pharmacological activities against human ailments including neurological diseases. In fact, tanshinones have been used to treat heart diseases, stroke, and vascular diseases in traditional Chinese medicine. During the last decade, tanshinones have been the most widely studied phytochemicals for their neuroprotective effects against experimental models of cerebral ischemia and Alzheimer's diseases. Importantly, tanshinone IIA, mostly studied tanshinone for biological activities, is recently reported to attenuate blood-brain barrier permeability among stroke patients, suggesting tanshinone IIA as an appealing therapeutic candidate for neurological diseases. Tanshinone I and IIA are also effective in experimental models of Parkinson's disease, Multiple sclerosis, and other neuroinflammatory diseases. In addition, several experimental studies suggested the pleiotropic neuroprotective effects of tanshinones such as anti-inflammatory, antioxidant, anti-apoptotic, and BBB protectant further value aiding to tanshinone as an appealing therapeutic strategy in neurological diseases. Therefore, in this review, we aimed to compile the recent updates and cellular and molecular mechanisms of neuroprotection of tanshinone IIA in diverse neurological diseases.
Collapse
Affiliation(s)
- Lalita Subedi
- Department of Anesthesiology and Neurology, Shock Trauma and Anesthesiology Research Center, University of Maryland, School of Medicine, Baltimore, MD, USA
| | - Bhakta Prasad Gaire
- Department of Anesthesiology and Neurology, Shock Trauma and Anesthesiology Research Center, University of Maryland, School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
14
|
Xu H, Wang E, Chen F, Xiao J, Wang M. Neuroprotective Phytochemicals in Experimental Ischemic Stroke: Mechanisms and Potential Clinical Applications. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6687386. [PMID: 34007405 PMCID: PMC8102108 DOI: 10.1155/2021/6687386] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/10/2021] [Accepted: 03/29/2021] [Indexed: 02/06/2023]
Abstract
Ischemic stroke is a challenging disease with high mortality and disability rates, causing a great economic and social burden worldwide. During ischemic stroke, ionic imbalance and excitotoxicity, oxidative stress, and inflammation are developed in a relatively certain order, which then activate the cell death pathways directly or indirectly via the promotion of organelle dysfunction. Neuroprotection, a therapy that is aimed at inhibiting this damaging cascade, is therefore an important therapeutic strategy for ischemic stroke. Notably, phytochemicals showed great neuroprotective potential in preclinical research via various strategies including modulation of calcium levels and antiexcitotoxicity, antioxidation, anti-inflammation and BBB protection, mitochondrial protection and antiapoptosis, autophagy/mitophagy regulation, and regulation of neurotrophin release. In this review, we summarize the research works that report the neuroprotective activity of phytochemicals in the past 10 years and discuss the neuroprotective mechanisms and potential clinical applications of 148 phytochemicals that belong to the categories of flavonoids, stilbenoids, other phenols, terpenoids, and alkaloids. Among them, scutellarin, pinocembrin, puerarin, hydroxysafflor yellow A, salvianolic acids, rosmarinic acid, borneol, bilobalide, ginkgolides, ginsenoside Rd, and vinpocetine show great potential in clinical ischemic stroke treatment. This review will serve as a powerful reference for the screening of phytochemicals with potential clinical applications in ischemic stroke or the synthesis of new neuroprotective agents that take phytochemicals as leading compounds.
Collapse
Affiliation(s)
- Hui Xu
- Institute for Advanced Study, Shenzhen University, Shenzhen 508060, China
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | | | - Feng Chen
- Institute for Advanced Study, Shenzhen University, Shenzhen 508060, China
| | - Jianbo Xiao
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
| | - Mingfu Wang
- Institute for Advanced Study, Shenzhen University, Shenzhen 508060, China
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| |
Collapse
|
15
|
Park Y, Ahn JH, Cho JH, Tae HJ, Lee TK, Kim B, Lee JC, Park JH, Shin MC, Ohk TG, Cho JH, Won MH. Effects of hypothermia on inflammatory cytokine expression in rat liver following asphyxial cardiac arrest. Exp Ther Med 2021; 21:626. [PMID: 33968162 PMCID: PMC8097226 DOI: 10.3892/etm.2021.10058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 03/17/2021] [Indexed: 12/18/2022] Open
Abstract
Hypothermic treatment is known to protect against cardiac arrest (CA) and improve survival rate. However, few studies have evaluated the CA-induced liver damage and the effects of hypothermia on this damage. Therefore, the aim of the present study was to determine possible protective effects of hypothermia on the liver after asphyxial CA. Rats were subjected to a 5-min asphyxial CA followed by return of spontaneous circulation (ROSC). The body temperature was controlled at 37±0.5˚C (normothermia group) or 33±0.5˚C (hypothermia group) for 4 h after ROSC. Livers were examined at 6, 12 h, 1 and 2 days after ROSC. Histopathological examination was performed by H&E staining. Alterations in the expression levels of pro-inflammatory (TNF-α and interleukin IL-2) and anti-inflammatory cytokines (IL-4 and IL-13) were investigated by immunohistochemistry. Sinusoidal dilatation and vacuolization were observed after asphyxial CA by histopathological examination. However, these CA-induced structural alterations were prevented by hypothermia. In immunohistochemical examination, the expression levels of pro-inflammatory cytokines were reduced in the hypothermia group compared with those in the normothermia group while the expression levels of anti-inflammatory cytokines were increased in the hypothermia group compared with those in the normothermia group. In conclusion, hypothermic treatment for 4 h following asphyxial CA in rats inhibited the increase of pro-inflammatory cytokines and stimulated the expression of anti-inflammatory cytokines compared with the normothermic group. The results of the present study suggested that hypothermic treatment after asphyxial CA reduced liver damage via the regulation of inflammation.
Collapse
Affiliation(s)
- Yoonsoo Park
- Department of Emergency Medicine, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24289, Republic of Korea
| | - Ji Hyeon Ahn
- Department of Physical Therapy, College of Health Science, Youngsan University, Yangsan, Gyeongnam 50510, Republic of Korea.,Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Jeong Hwi Cho
- Bio-Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeollabuk 54596, Republic of Korea
| | - Hyun-Jin Tae
- Bio-Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeollabuk 54596, Republic of Korea
| | - Tae-Kyeong Lee
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Bora Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Joon Ha Park
- Department of Anatomy, College of Korean Medicine, Dongguk University, Gyeongju, Gyeongbuk 38066, Republic of Korea
| | - Myoung Cheol Shin
- Department of Emergency Medicine, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24289, Republic of Korea
| | - Taek Geun Ohk
- Department of Emergency Medicine, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24289, Republic of Korea
| | - Jun Hwi Cho
- Department of Emergency Medicine, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24289, Republic of Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| |
Collapse
|
16
|
Wang Z, He C, Shi JS. Natural Products for the Treatment of Neurodegenerative Diseases. Curr Med Chem 2020; 27:5790-5828. [PMID: 31131744 DOI: 10.2174/0929867326666190527120614] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/06/2019] [Accepted: 05/09/2019] [Indexed: 12/13/2022]
Abstract
Neurodegenerative diseases are a heterogeneous group of disorders characterized by the progressive degeneration of the structure and function of the central nervous system or peripheral nervous system. Alzheimer's Disease (AD), Parkinson's Disease (PD) and Spinal Cord Injury (SCI) are the common neurodegenerative diseases, which typically occur in people over the age of 60. With the rapid development of an aged society, over 60 million people worldwide are suffering from these uncurable diseases. Therefore, the search for new drugs and therapeutic methods has become an increasingly important research topic. Natural products especially those from the Traditional Chinese Medicines (TCMs), are the most important sources of drugs, and have received extensive interest among pharmacist. In this review, in order to facilitate further chemical modification of those useful natural products by pharmacists, we will bring together recent studies in single natural compound from TCMs with neuroprotective effect.
Collapse
Affiliation(s)
- Ze Wang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi Guizhou 563003, China.,Generic Drug Research Center of Guizhou Province, School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, 563003, P.R. China
| | - Chunyang He
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi Guizhou 563003, China.,Generic Drug Research Center of Guizhou Province, School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, 563003, P.R. China
| | - Jing-Shan Shi
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi Guizhou 563003, China
| |
Collapse
|
17
|
Park Y, Ahn JH, Lee TK, Kim B, Tae HJ, Park JH, Shin MC, Cho JH, Won MH. Therapeutic hypothermia reduces inflammation and oxidative stress in the liver after asphyxial cardiac arrest in rats. Acute Crit Care 2020; 35:286-295. [PMID: 33423440 PMCID: PMC7808856 DOI: 10.4266/acc.2020.00304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 11/12/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Few studies have evaluated the effects of hypothermia on cardiac arrest (CA)-induced liver damage. This study aimed to investigate the effects of hypothermic therapy on the liver in a rat model of asphyxial cardiac arrest (ACA). METHODS Rats were subjected to 5-minute ACA followed by return of spontaneous circulation (RoSC). Body temperature was controlled at 33°C±0.5°C or 37°C±0.5°C for 4 hours after RoSC in the hypothermia group and normothermia group, respectively. Liver tissues in each group were collected at 6 hours, 12 hours, 1 day, and 2 days after RoSC. To examine hepatic inflammation, mast cells were stained with toluidine blue. Superoxide anion radical production was evaluated using dihydroethidium fluorescence straining and expression of endogenous antioxidants (superoxide dismutase 1 [SOD1] and SOD2) was examined using immunohistochemistry. RESULTS There were significantly more mast cells in the livers of the normothermia group with ACA than in the hypothermia group with ACA. Gradual increase in superoxide anion radical production was found with time in the normothermia group with ACA, but production was significantly suppressed in the hypothermia group with ACA relative to the normothermia group with ACA. SOD1 and SOD2 levels were higher in the hypothermia group with ACA than in the normothermia group with ACA. CONCLUSIONS Experimental hypothermic treatment after ACA significantly inhibited inflammation and superoxide anion radical production in the rat liver, indicating that this treatment enhanced or maintained expression of antioxidants. Our findings suggest that hypothermic therapy after CA can reduce mast cell-mediated inflammation through regulation of oxidative stress and the expression of antioxidants in the liver.
Collapse
Affiliation(s)
- Yoonsoo Park
- Department of Emergency Medicine, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon, Korea
| | - Ji Hyeon Ahn
- Department of Physical Therapy, College of Health Science, Youngsan University, Yangsan, Korea.,Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Korea
| | - Tae-Kyeong Lee
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, Korea
| | - Bora Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Korea
| | - Hyun-Jin Tae
- Bio-Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Korea
| | - Joon Ha Park
- Department of Anatomy, College of Korean Medicine, Dongguk University, Gyeongju, Korea
| | - Myoung Cheol Shin
- Department of Emergency Medicine, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon, Korea
| | - Jun Hwi Cho
- Department of Emergency Medicine, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon, Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Korea
| |
Collapse
|
18
|
Foulkes MJ, Tolliday FH, Henry KM, Renshaw SA, Jones S. Evaluation of the anti-inflammatory effects of synthesised tanshinone I and isotanshinone I analogues in zebrafish. PLoS One 2020; 15:e0240231. [PMID: 33022012 PMCID: PMC7537861 DOI: 10.1371/journal.pone.0240231] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 09/22/2020] [Indexed: 01/13/2023] Open
Abstract
During inflammation, dysregulated neutrophil behaviour can play a major role in a range of chronic inflammatory diseases, for many of which current treatments are generally ineffective. Recently, specific naturally occurring tanshinones have shown promising anti-inflammatory effects by targeting neutrophils in vivo, yet such tanshinones, and moreover, their isomeric isotanshinone counterparts, are still a largely underexplored class of compounds, both in terms of synthesis and biological effects. To explore the anti-inflammatory effects of isotanshinones, and the tanshinones more generally, a series of substituted tanshinone and isotanshinone analogues was synthesised, alongside other structurally similar molecules. Evaluation of these using a transgenic zebrafish model of neutrophilic inflammation revealed differential anti-inflammatory profiles in vivo, with a number of compounds exhibiting promising effects. Several compounds reduce initial neutrophil recruitment and/or promote resolution of neutrophilic inflammation, of which two also result in increased apoptosis of human neutrophils. In particular, the methoxy-substituted tanshinone 39 specifically accelerates resolution of inflammation without affecting the recruitment of neutrophils to inflammatory sites, making this a particularly attractive candidate for potential pro-resolution therapeutics, as well as a possible lead for future development of functionalised tanshinones as molecular tools and/or chemical probes. The structurally related β-lapachones promote neutrophil recruitment but do not affect resolution. We also observed notable differences in toxicity profiles between compound classes. Overall, we provide new insights into the in vivo anti-inflammatory activities of several novel tanshinones, isotanshinones, and structurally related compounds.
Collapse
Affiliation(s)
- Matthew J. Foulkes
- Department of Chemistry, The University of Sheffield, Sheffield, United Kingdom
- The Bateson Centre, The University of Sheffield, Sheffield, United Kingdom
- Department of Infection, Immunity & Cardiovascular Disease, The University of Sheffield, Sheffield, United Kingdom
| | - Faith H. Tolliday
- The Bateson Centre, The University of Sheffield, Sheffield, United Kingdom
- Department of Infection, Immunity & Cardiovascular Disease, The University of Sheffield, Sheffield, United Kingdom
| | - Katherine M. Henry
- The Bateson Centre, The University of Sheffield, Sheffield, United Kingdom
- Department of Infection, Immunity & Cardiovascular Disease, The University of Sheffield, Sheffield, United Kingdom
| | - Stephen A. Renshaw
- The Bateson Centre, The University of Sheffield, Sheffield, United Kingdom
- Department of Infection, Immunity & Cardiovascular Disease, The University of Sheffield, Sheffield, United Kingdom
| | - Simon Jones
- Department of Chemistry, The University of Sheffield, Sheffield, United Kingdom
- * E-mail:
| |
Collapse
|
19
|
Pharmacological basis of tanshinone and new insights into tanshinone as a multitarget natural product for multifaceted diseases. Biomed Pharmacother 2020; 130:110599. [PMID: 33236719 DOI: 10.1016/j.biopha.2020.110599] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 07/18/2020] [Accepted: 07/31/2020] [Indexed: 12/14/2022] Open
Abstract
Drug development has long included the systematic exploration of various resources. Among these, natural products are one of the most important resources from which novel agents are developed due to the multiple pharmacologic effects of these natural products on diseases. Tanshinone, a representative natural product, is the main compound extracted from the dried root and rhizome of Salvia miltiorrhiza Bge. Research on tanshinone began in the early 1930s. With the in-depth investigation of an increasing number of identified analogs, tanshinone has demonstrated a wide variety of bioactivities and contradicted the saying, 'You can't teach an old dog new tricks'. This review is focused on the pharmacological action of tanshinone and status of research on tanshinone in recent years. The mechanism of tanshinone has also drawn much attention, with the findings of representative targets and pathways of tanshinone. The most recent studies have comprehensively shown that tanshinone can be used to treat leukemia and solid carcinoma, protect against cardiovascular and cerebrovascular diseases, and alleviate liver- and kidney-related diseases, among its other effects. Multiple signaling pathways, including antiproliferative, antiapoptotic, anti-inflammatory, and antioxidative stress pathways, are involved in its actions.
Collapse
|
20
|
Experimental Pretreatment with Chlorogenic Acid Prevents Transient Ischemia-Induced Cognitive Decline and Neuronal Damage in the Hippocampus through Anti-Oxidative and Anti-Inflammatory Effects. Molecules 2020; 25:molecules25163578. [PMID: 32781658 PMCID: PMC7463954 DOI: 10.3390/molecules25163578] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/03/2020] [Accepted: 08/05/2020] [Indexed: 12/17/2022] Open
Abstract
Chlorogenic acid (CGA), an ester of caffeic acid and quinic acid, is among the phenolic acid compounds which can be naturally found in green coffee extract and tea. CGA has been studied since it displays significant pharmacological properties. The aim of this study was to investigate the effects of CGA on cognitive function and neuroprotection including its mechanisms in the hippocampus following transient forebrain ischemia in gerbils. Memory and learning following the ischemia was investigated by eight-arm radial maze and passive avoidance tests. Neuroprotection was examined by immunohistochemistry for neuronal nuclei-specific protein and Fluoro-Jade B histofluorescence staining. For mechanisms of the neuroprotection, alterations in copper, zinc-superoxide dismutase (SOD1), SOD2 as antioxidant enzymes, dihydroethidium and 4-hydroxy-2-nonenal as indicators for oxidative stress, and anti-inflammatory cytokines (interleukin (IL)-4 and IL-13) and pro-inflammatory cytokines (tumor necrosis factor α (TNF-α) and IL-2) were examined by Western blotting and/or immunohistochemistry. As a result, pretreatment with 30 mg/kg CGA attenuated cognitive impairment and displayed a neuroprotective effect against transient forebrain ischemia (TFI). In Western blotting, the expression levels of SOD2 and IL-4 were increased due to pretreatment with CGA and, furthermore, 4-HNE production and IL-4 expressions were inhibited by CGA pretreatment. Additionally, pretreated CGA enhanced antioxidant enzymes and anti-inflammatory cytokines and, in contrast, attenuated oxidative stress and pro-inflammatory cytokine expression. Based on these results, we suggest that CGA can be a useful neuroprotective material against ischemia-reperfusion injury due to its antioxidant and anti-inflammatory efficacies.
Collapse
|
21
|
Lee TK, Park Y, Kim B, Lee JC, Shin MC, Ohk TG, Park CW, Cho JH, Park JH, Lee CH, Won MH, Ahn JH. Long-Term Alternating Fasting Increases Interleukin-13 in the Gerbil Hippocampus, But Does Not Protect BBB and Pyramidal Neurons From Ischemia-Reperfusion Injury. Neurochem Res 2020; 45:2352-2363. [PMID: 32671629 DOI: 10.1007/s11064-020-03094-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 07/07/2020] [Accepted: 07/08/2020] [Indexed: 12/11/2022]
Abstract
It is questionable whether intermittent fasting (IF) protects against brain ischemic injury. This study examined whether IF increased anti-inflammatory cytokines and protected neurons from ischemia-reperfusion injury in the gerbil hippocampus. Gerbils were subjected to 1-day alternating fasting as IF for 1, 2, or 3 months and assigned to sham or 5 min of transient ischemia. We examined the changes in anti-inflammatory cytokines (IL-4 and IL-13), neurons and IgG by immunohistochemistry or immunofluorescence staining in the cornu ammonis 1 (CA1) region of the hippocampus before and after ischemia. IF increased IL-13 immunoreactivity in the CA1 region before ischemia, but did not affect IL-4 immunoreactivity. After ischemia, IL-13 and 4 immunoreactivities in the CA1 region were significantly lower in IF gerbils than in non-IF gerbils. In the IF gerbils, the CA1 pyramidal neurons were not protected from ischemic injury; in these gerbils, strong IgG immunoreactivity was seen in the CA1 parenchyma, indicating leakage of the BBB. In brief, IF increased IL-13 in the CA1 region, but these neurons were not protected from transient ischemic injury evidenced by IgG immunoreactivity in the CA1 parenchyma. This study indicates that IF increased some anti-inflammatory cytokines but did not afford neuroprotection against ischemic insults via BBB disruption.
Collapse
Affiliation(s)
- Tae-Kyeong Lee
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon, 24252, Republic of Korea
| | - Yoonsoo Park
- Department of Emergency Medicine, and Institute of Medical Sciences, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Bora Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Myoung Cheol Shin
- Department of Emergency Medicine, and Institute of Medical Sciences, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Taek Geun Ohk
- Department of Emergency Medicine, and Institute of Medical Sciences, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Chan Woo Park
- Department of Emergency Medicine, and Institute of Medical Sciences, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Jun Hwi Cho
- Department of Emergency Medicine, and Institute of Medical Sciences, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Joon Ha Park
- Department of Anatomy, College of Oriental Medicine, Dongguk University-Gyeongju, Gyeongju, Gyeongbuk, 38066, Republic of Korea
| | - Choong Hyun Lee
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan, Chungnam, 31116, Republic of Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea.
| | - Ji Hyeon Ahn
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon, 24252, Republic of Korea.
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea.
| |
Collapse
|
22
|
Park JH, Lee TK, Kim DW, Park CW, Park YE, Kim B, Lee JC, Lee HA, Won MH, Ahn JH. RbAp48 expression and neuronal damage in the gerbil hippocampus following 5 min of transient ischemia. Lab Anim Res 2020; 35:12. [PMID: 32257900 PMCID: PMC7081550 DOI: 10.1186/s42826-019-0011-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Accepted: 07/14/2019] [Indexed: 11/10/2022] Open
Abstract
Histone-binding protein RbAp48 has been known to be involved in histone acetylation, and epigenetic alterations of histone modifications are closely associated with the pathogenesis of ischemic reperfusion injury. In the current study, we investigated chronological change of RbAp48 expression in the hippocampus following 5 min of transient ischemia in gerbils. RbAp48 expression was examined 1, 2, 5, and 10 days after transient ischemia using immunohistochemistry. In sham operated gerbils, RbAp48 immunoreactivity was strong in pyramidal and non-pyramidal cells in the hippocampus. After transient ischemia, RbAp48 immunoreactivity was changed in the cornu ammonis 1 subfield (CA1), not in CA2/3. RbAp48 immunoreactivity in CA1 pyramidal neurons was gradually decreased and not detected at 5 and 10 days after ischemia. RbAp48 immunoreactivity in non-pyramidal cells was maintained until 2 days post-ischemia and significantly increased from 5 days post-ischemia. Double immunohistofluorescence staining revealed that RbAp48 immunoreactive non-pyramidal cells were astrocytes. At 5 days post-ischemia, death of pyramidal neurons occurred only in the CA1. These results showed that RbAp48 immunoreactivity was distinctively altered in pyramidal neurons and astrocytes in the hippocampal CA1 following 5 mins of transient ischemia. Ischemia-induced change in RbAp48 expression may be closely associated with neuronal death and astrocyte activation following 5 min of transient ischemia.
Collapse
Affiliation(s)
- Joon Ha Park
- 1Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252 Republic of Korea
| | - Tae-Kyeong Lee
- 2Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341 Republic of Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, and Research Institute of Oral Sciences, College of Dentistry, Gangnung-Wonju National University, Gangneung, Gangwon 25457 Republic of Korea
| | - Cheol Woo Park
- 2Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341 Republic of Korea
| | - Young Eun Park
- 2Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341 Republic of Korea
| | - Bora Kim
- 2Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341 Republic of Korea
| | - Jae-Chul Lee
- 2Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341 Republic of Korea
| | - Hyang-Ah Lee
- 4Department of Obstetrics and Gynecology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341 Republic of Korea
| | - Moo-Ho Won
- 2Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341 Republic of Korea
| | - Ji Hyeon Ahn
- 1Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252 Republic of Korea
| |
Collapse
|
23
|
Kim W, Kang MS, Kim TH, Yoo DY, Park JH, Jung HY, Won MH, Choi JH, Hwang IK. Ischemia-related changes of fat-mass and obesity-associated protein expression in the gerbil hippocampus. Metab Brain Dis 2020; 35:335-342. [PMID: 31786728 DOI: 10.1007/s11011-019-00513-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 11/05/2019] [Indexed: 01/05/2023]
Abstract
Fat-mass and obesity-associated protein (Fto) plays important roles in energy metabolism. It also acts as a demethylase and is most abundantly found in the brain. In the present study, we examined the spatial and temporal changes of Fto immunoreactivity after five minutes of transient forebrain ischemia in the hippocampus. In the control group, Fto immunoreactivity was mainly observed in the nucleus of pyramidal cells in the CA1 and CA3 regions as well as the polymorphic layer, granule cell layer, and subgranular zone of the dentate gyrus. Fto immunoreactivity was transiently, but not significantly, increased in the hippocampal CA3 region and the dentate gyrus two days after ischemia compared to mice without ischemia in the sham-operated group. Four days after ischemia, low Fto immunoreactivity was observed in the stratum pyramidale of the CA1 region because of neuronal death, but Fto immunoreactive cells were abundantly detected in the stratum pyramidale of the CA3 region, which is relatively resistant to ischemic damage. Thereafter, Fto immunoreactivity progressively decreased in the hippocampal CA1 and CA3 regions and the dentate gyrus until ten days after ischemia. At this time-point, Fto immunoreactivity was significantly lower in the hippocampal CA1 and CA3 regions and the dentate gyrus compared to that in the sham-operated group. The reduction of Fto immunoreactive structures in the hippocampus may be associated with impairments in Fto-related hippocampal function.
Collapse
Affiliation(s)
- Woosuk Kim
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, 85-508 Seoul National University, 1 Gwanak-ro, Seoul, 08826, South Korea
- Department of Biomedical Sciences, and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Min Soo Kang
- Department of Anatomy, College of Veterinary Medicine and Institute of Veterinary Science, 411-105 Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, 24341, South Korea
| | - Tae Hyeong Kim
- Department of Anatomy, College of Veterinary Medicine and Institute of Veterinary Science, 411-105 Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, 24341, South Korea
| | - Dae Young Yoo
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan, 31151, South Korea
| | - Joon Ha Park
- Department of Anatomy, College of Korean Medicine,, Dongguk University, Gyeongju, 38066, Republic of Korea
| | - Hyo Young Jung
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, 85-508 Seoul National University, 1 Gwanak-ro, Seoul, 08826, South Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 24341, South Korea
| | - Jung Hoon Choi
- Department of Anatomy, College of Veterinary Medicine and Institute of Veterinary Science, 411-105 Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, 24341, South Korea.
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, 85-508 Seoul National University, 1 Gwanak-ro, Seoul, 08826, South Korea.
| |
Collapse
|
24
|
Wang X, Yang Y, Liu X, Gao X. Pharmacological properties of tanshinones, the natural products from Salvia miltiorrhiza. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2020; 87:43-70. [PMID: 32089238 DOI: 10.1016/bs.apha.2019.10.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Danshen (Cai, et al. 2016) is the dry root and rhizome of the herbaceous plant Salvia miltiorrhiza Bge. of family labiatae, a perennial plant that is native to China and Japan. The primary modern clinical applications of Danshen are for heart disease, chronic hepatitis, early cirrhosis, cerebral ischemia and pulmonary heart disease. Emerging evidence from cellular, animal, and clinical studies has begun to illuminate the pharmacological attributes of the primary lipophilic tanshinones from Danshen, which include tanshinone I, tanshinone II, cryptotanshinone and dihydrotanshinone, etc. Tanshinones offer the properties of anti-oxidation, anti-inflammation, antitumor, phytoestrogenic activity, vasodilation, neuroprotection, regulate metabolic function and other pharmacological advances. This chapter will review the discovery of the pharmacodynamic mechanism and pharmacokinetic studies of tanshinones and Danshen for further clinical applications.
Collapse
Affiliation(s)
- Xiaoying Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yang Yang
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiao Liu
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiumei Gao
- Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| |
Collapse
|
25
|
Im JH, Yeo IJ, Park PH, Choi DY, Han SB, Yun J, Hong JT. Deletion of Chitinase-3-like 1 accelerates stroke development through enhancement of Neuroinflammation by STAT6-dependent M2 microglial inactivation in Chitinase-3-like 1 knockout mice. Exp Neurol 2020; 323:113082. [PMID: 31669069 DOI: 10.1016/j.expneurol.2019.113082] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/02/2019] [Accepted: 10/12/2019] [Indexed: 11/18/2022]
Abstract
Chitinase 3-like 1 (Chi3L1) plays a major role in the pathogenesis of inflammatory diseases. We investigated the effect of Chi3L1 knockout on stroke development. Ischemia/reperfusion was induced by middle cerebral artery occlusion (MCAO) in Chi3L1 knockout and wildtype mice. Significantly increased infarct volume and decreased neurological deficit scores at 24 h after ischemia/reperfusion were found in Chi3L1 knockout mice compared to wildtype mice. Moreover, ischemic neuronal cell death was increased in Chi3L1 knockout mice through increased oxidative stress and release of IL-6 and IL-1β but IL-10 and IL-4 were reduced. Furthermore, expression of inflammation-related proteins (iNOS, COX-2, Iba-1, and GFAP) was significantly increased in Chi3L1 knockout mice compared to wildtype. In microglia isolated from MCAO-injured Chi3L1 knockout mice, expression of M1 markers (iNOS, CD86, IL-1β, and IL-6) was increased and M2 markers (Arg1, Mrc1, IL-10, and IL-4Ra) was decreased. In BV-2 cells, knockdown of Chi3L1 increased TNF-α- and INF-γ-induced expression of iNOS, COX-2, and Iba-1, but decreased the expression of Arg1, MRC1, and IL-4 receptor-alpha (IL-4Rα). Expression of IL-4Rα, an important factor of M2 polarization, and its downstream signals p-JAK1, p-JAK3, and p-STAT6, was much reduced in the knockout mice. Additionally, in BV-2 cells, knockdown of Chi3L1 by siRNA Chi3L1 decreased rhTNF-α- and INF-γ-induced expression of IL-4Rα, p-JAK1, p-JAK3, and p-STAT6. Furthermore, treatment with AS1517499 abolished Chi3L1 knockdown-induced reduced IL-4Rα and Arg1 but not CD86 expression. Our results indicate that deletion of Chi3L1 accelerates stroke development through enhancement of neuroinflammation by markedly decreasing STAT6-dependent M2 macrophage polarization.
Collapse
Affiliation(s)
- Jun Hyung Im
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-21, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do, Republic of Korea
| | - In Jun Yeo
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-21, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do, Republic of Korea
| | - Pil Hoon Park
- College of Pharmacy, Yeungnam University, 280 Daehak-Ro, Gyeongsan, Gyeongbuk, Republic of Korea
| | - Dong Young Choi
- College of Pharmacy, Yeungnam University, 280 Daehak-Ro, Gyeongsan, Gyeongbuk, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-21, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do, Republic of Korea
| | - Jaesuk Yun
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-21, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-21, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do, Republic of Korea.
| |
Collapse
|
26
|
Chang P, Tian Y, Williams AM, Bhatti UF, Liu B, Li Y, Alam HB. Inhibition of Histone Deacetylase 6 Protects Hippocampal Cells Against Mitochondria-mediated Apoptosis in a Model of Severe Oxygen-glucose Deprivation. Curr Mol Med 2019; 19:673-682. [DOI: 10.2174/1566524019666190724102755] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 07/05/2019] [Accepted: 07/08/2019] [Indexed: 11/22/2022]
Abstract
Background:
Histone deacetylase (HDAC) 6 inhibitors have demonstrated
significant protective effects in traumatic injuries. However, their roles in neuroprotection
and underlying mechanisms are poorly understood. This study sought to investigate the
neuroprotective effects of Tubastatin A (Tub-A), an HDAC6 inhibitor, during oxygenglucose
deprivation (OGD) in HT22 hippocampal cells.
Methods:
HT22 hippocampal cells were exposed to OGD. Cell viability and cytotoxicity
were assessed by cell counting kit-8 (CCK-8) and lactate dehydrogenase (LDH) release
assay. Cellular apoptosis was assessed by Terminal deoxynucleotidyl transferase dUTP
nick end labeling (TUNEL) assay. Mitochondria membrane potential was detected using
JC-1 dye. Expressions of acetylated α-tubulin, α-tubulin, cytochrome c, VDAC, Bax, Bcl-
2, cleaved caspase 3, phosphorylated Akt, Akt, phosphorylated GSK3β and GSK3β
were analyzed by Western blot analysis.
Results:
Tub-A induced acetylation of α-tubulin, demonstrating appropriate efficacy.
Tub-A significantly increased cell viability and attenuated LDH release after exposure to
OGD. Furthermore, Tub-A treatment blunted the increase in TUNEL-positive cells
following OGD and preserved the mitochondrial membrane potential. Tub-A also
attenuated the release of cytochrome c from the mitochondria into the cytoplasm and
suppressed the ratio of Bax/Bcl-2 and cleaved caspase 3. This was mediated, in part, by
the increased phosphorylation of Akt and GSK3β signaling pathways.
Conclusion:
HDAC 6 inhibition, using Tub-A, protects against OGD-induced injury in
HT22 cells by modulating Akt/GSK3β signaling and inhibiting mitochondria-mediated
apoptosis.
Collapse
Affiliation(s)
- Panpan Chang
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, United States
| | - Yuzi Tian
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, United States
| | - Aaron M. Williams
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, United States
| | - Umar F. Bhatti
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, United States
| | - Baoling Liu
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, United States
| | - Yongqing Li
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, United States
| | - Hasan B. Alam
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, United States
| |
Collapse
|
27
|
Zhang KJ, Zheng Q, Zhu PC, Tong Q, Zhuang Z, Zhu JZ, Bao XY, Huang YY, Zheng GQ, Wang Y. Traditional Chinese Medicine for Coronary Heart Disease: Clinical Evidence and Possible Mechanisms. Front Pharmacol 2019; 10:844. [PMID: 31427964 PMCID: PMC6688122 DOI: 10.3389/fphar.2019.00844] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 07/01/2019] [Indexed: 12/23/2022] Open
Abstract
Coronary heart disease (CHD) remains a major cause of mortality with a huge economic burden on healthcare worldwide. Here, we conducted a systematic review to investigate the efficacy and safety of Chinese herbal medicine (CHM) for CHD based on high-quality randomized controlled trials (RCTs) and summarized its possible mechanisms according to animal-based researches. 27 eligible studies were identified in eight database searches from inception to June 2018. The methodological quality was assessed using seven-item checklist recommended by Cochrane Collaboration. All the data were analyzed using Rev-Man 5.3 software. As a result, the score of study quality ranged from 4 to 7 points. Meta-analyses showed CHM can significantly reduce the incidence of myocardial infarction and percutaneous coronary intervention, and cardiovascular mortality (P < 0.05), and increase systolic function of heart, the ST-segment depression, and clinical efficacy (P < 0.05). Adverse events were reported in 11 studies, and CHMs were well tolerated in patients with CHD. In addition, CHM exerted cardioprotection for CHD, possibly altering multiple signal pathways through anti-inflammatory, anti-oxidation, anti-apoptosis, improving the circulation, and regulating energy metabolism. In conclusion, the evidence available from present study revealed that CHMs are beneficial for CHD and are generally safe.
Collapse
Affiliation(s)
- Ke-Jian Zhang
- Department of Cardiology, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qun Zheng
- Department of Cardiology, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Peng-Chong Zhu
- Department of Cardiology, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qiang Tong
- Department of Cardiology, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhuang Zhuang
- Department of Cardiology, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jia-Zhen Zhu
- Department of Cardiology, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiao-Yi Bao
- Department of Cardiology, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yue-Yue Huang
- Department of Cardiology, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Guo-Qing Zheng
- Department of Neurology, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yan Wang
- Department of Cardiology, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
28
|
Ahn JH, Kim DW, Park JH, Lee TK, Lee HA, Won MH, Lee CH. Expression changes of CX3CL1 and CX3CR1 proteins in the hippocampal CA1 field of the gerbil following transient global cerebral ischemia. Int J Mol Med 2019; 44:939-948. [PMID: 31524247 PMCID: PMC6658004 DOI: 10.3892/ijmm.2019.4273] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 07/08/2019] [Indexed: 01/27/2023] Open
Abstract
Chemokine C-X3-C motif ligand 1 (CX3CL1) and its sole receptor, CX3CR1, are known to be involved in neuronal damage/death following brain ischemia. In the present study, time-dependent expression changes of CX3CL1 and CX3CR1 proteins were investigated in the hippocampal CA1 field following 5 min of transient global cerebral ischemia (tgCI) in gerbils. To induce tgCI in gerbils, bilateral common carotid arteries were occluded for 5 min using aneurysm clips. Expression changes of CX3CL1 and CX3CR1 proteins were assessed at 1, 2 and 5 days after tgCI using western blotting and immunohistochemistry. CX3CL1 immunoreactivity was strong in the CA1 pyramidal cells of animals in the sham operation group. Weak CX3CL1 immunoreactivity was detected at 6 h after tgCI, recovered at 1 day after tgCI and disappeared from 5 days after tgCI. CX3CR1 immunoreactivity was very weak in CA1 pyramidal cells of the sham animals. CX3CR1 immunoreactivity in CA1 pyramidal cells was significantly increased at 1 days after tgCI and gradually decreased thereafter. On the other hand, CX3CR1 immunoreactivity was significantly increased in microglia from 5 days after tgCI. These results showed that CX3CL1 and CX3CR1 protein expression levels in pyramidal cells and microglia in the hippocampal CA1 field following tgCI were changed, indicating that tgCI-induced expression changes of CX3CL1 and CX3CR1 proteins might be closely associated with tgCI-induced delayed neuronal death and microglial activation.
Collapse
Affiliation(s)
- Ji Hyeon Ahn
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, and Research Institute of Oral Sciences, College of Dentistry, Gangnung‑Wonju National University, Gangneung, Gangwon 25457, Republic of Korea
| | - Joon Ha Park
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Tae-Kyeong Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Hyang-Ah Lee
- Department of Obstetrics and Gynecology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Choong-Hyun Lee
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan, Chungcheongnam 31116, Republic of Korea
| |
Collapse
|
29
|
Wu Y, Zhang B, Kebebe D, Guo L, Guo H, Li N, Pi J, Qi D, Guo P, Liu Z. Preparation, optimization and cellular uptake study of tanshinone I nanoemulsion modified with lactoferrin for brain drug delivery. Pharm Dev Technol 2019; 24:982-991. [DOI: 10.1080/10837450.2019.1621897] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Yumei Wu
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Bing Zhang
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Dereje Kebebe
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
- School of Pharmacy, Institute of Health Sciences, Jimma University, Jimma, Ethiopia
| | - Lili Guo
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Hong Guo
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Nan Li
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Jiaxin Pi
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Dongli Qi
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Pan Guo
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Zhidong Liu
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| |
Collapse
|
30
|
Wu YT, Bi YM, Tan ZB, Xie LP, Xu HL, Fan HJ, Chen HM, Li J, Liu B, Zhou YC. Tanshinone I inhibits vascular smooth muscle cell proliferation by targeting insulin-like growth factor-1 receptor/phosphatidylinositol-3-kinase signaling pathway. Eur J Pharmacol 2019; 853:93-102. [PMID: 30878387 DOI: 10.1016/j.ejphar.2019.03.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 03/03/2019] [Accepted: 03/12/2019] [Indexed: 02/07/2023]
Abstract
Vascular smooth muscle cell (VSMC) proliferation plays a critical role in arterial remodeling during various vascular diseases including atherosclerosis and hypertension. Tanshinone I, a major component of Salvia miltiorrhiza, exerts protective effects against cardiovascular diseases. In this study, we investigated the effects of tanshinone I on VSMC proliferation, as well as the underlying mechanisms. We found that this compound inhibited the proliferation of VSMCs in a dose-dependent manner, based on 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) and 5-ethynyl-2'-deoxyuridine (EdU) assays. Western blotting demonstrated that tanshinone I inhibited the expression of proliferation-related proteins, including cyclin-dependent kinase 4 (CDK4), cyclin D3, and cyclin D1, in a dose-dependent manner. Molecular docking showed that this compound docked to the inhibitor-binding site of the insulin-like growth factor 1 (IGF-1) receptor (IGF-1R), and the binding energy between tanshinone I and IGF-1R was -9.021 kcal/mol. Molecular dynamic simulations showed that the IGF-1R-tanshinone I binding was stable. We also found that tanshinone I dose-dependently inhibited IGF-1R activation and its downstream molecules, insulin receptor substrate (IRS)-1, phosphatidylinositol-3-Kinase (PI3K), Akt, glycogen synthase kinase-3 beta (GSK3β), mammalian target of rapamycin (mTOR), 70S6K, and ribosomal protein S6 (RPS6). Notably, activation of IGF-1R by recombinant IGF-1 rescued the activity of IGF-1R and its downstream molecules, and the proliferation of tanshinone I-treated VSMC. In addition, blocking PI3K signaling with LY294002 showed the important role of this pathway in tanshinone I-mediated suppression of VSMC proliferation. Collectively, these data demonstrated that tanshinone I might inhibit VSMC proliferation by inhibiting IGF-1R/PI3K signaling.
Collapse
Affiliation(s)
- Yu-Ting Wu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China; Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yi-Ming Bi
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China; Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhang-Bin Tan
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China; Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ling-Peng Xie
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China; Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Hong-Lin Xu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China; Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Hui-Jie Fan
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China; Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Hong-Mei Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China; Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jun Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China; Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Bin Liu
- Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Guangzhou Institute of Cardiovascular Disease, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China.
| | - Ying-Chun Zhou
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China; Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
31
|
Peng T, Jiang Y, Farhan M, Lazarovici P, Chen L, Zheng W. Anti-inflammatory Effects of Traditional Chinese Medicines on Preclinical in vivo Models of Brain Ischemia-Reperfusion-Injury: Prospects for Neuroprotective Drug Discovery and Therapy. Front Pharmacol 2019; 10:204. [PMID: 30930774 PMCID: PMC6423897 DOI: 10.3389/fphar.2019.00204] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 02/18/2019] [Indexed: 12/28/2022] Open
Abstract
Acquired brain ischemia-and reperfusion-injury (IRI), including both Ischemic stroke (IS) and Traumatic Brain injury (TBI), is one of the most common causes of disability and death in adults and represents a major burden in both western and developing countries worldwide. China’s clinical neurological therapeutic experience in the use of traditional Chinese medicines (TCMs), including TCM-derived active compounds, Chinese herbs, TCM formulations and decoction, in brain IRI diseases indicated a trend of significant improvement in patients’ neurological deficits, calling for blind, placebo-controlled and randomized clinical trials with careful meta-analysis evaluation. There are many TCMs in use for brain IRI therapy in China with significant therapeutic effects in preclinical studies using different brain IRI-animal. The basic hypothesis in this field claims that in order to avoid the toxicity and side effects of the complex TCM formulas, individual isolated and identified compounds that exhibited neuroprotective properties could be used as lead compounds for the development of novel drugs. China’s efforts in promoting TCMs have contributed to an explosive growth of the preclinical research dedicated to the isolation and identification of TCM-derived neuroprotective lead compounds. Tanshinone, is a typical example of TCM-derived lead compounds conferring neuroprotection toward IRI in animals with brain middle cerebral artery occlusion (MCAO) or TBI models. Recent reports show the significance of the inflammatory response accompanying brain IRI. This response appears to contribute to both primary and secondary ischemic pathology, and therefore anti-inflammatory strategies have become popular by targeting pro-inflammatory and anti-inflammatory cytokines, other inflammatory mediators, reactive oxygen species, nitric oxide, and several transcriptional factors. Here, we review recent selected studies and discuss further considerations for critical reevaluation of the neuroprotection hypothesis of TCMs in IRI therapy. Moreover, we will emphasize several TCM’s mechanisms of action and attempt to address the most promising compounds and the obstacles to be overcome before they will enter the clinic for IRI therapy. We hope that this review will further help in investigations of neuroprotective effects of novel molecular entities isolated from Chinese herbal medicines and will stimulate performance of clinical trials of Chinese herbal medicine-derived drugs in IRI patients.
Collapse
Affiliation(s)
- Tangming Peng
- Center of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau, China.,Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Macau, China.,Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China.,Neurosurgical Clinical Research Center of Sichuan Province, Luzhou, China
| | - Yizhou Jiang
- Center of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau, China.,Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Macau, China
| | - Mohd Farhan
- Center of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau, China.,Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Macau, China
| | - Philip Lazarovici
- Faculty of Medicine, School of Pharmacy, The Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ligang Chen
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China.,Neurosurgical Clinical Research Center of Sichuan Province, Luzhou, China
| | - Wenhua Zheng
- Center of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau, China.,Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Macau, China
| |
Collapse
|
32
|
Park JH, Kim IH, Ahn JH, Noh YH, Kim SS, Lee TK, Lee JC, Shin BN, Sim TH, Lee HS, Cho JH, Hwang IK, Kang IJ, Kim JD, Won MH. Pretreated Oenanthe Javanica extract increases anti-inflammatory cytokines, attenuates gliosis, and protects hippocampal neurons following transient global cerebral ischemia in gerbils. Neural Regen Res 2019; 14:1536-1543. [PMID: 31089052 PMCID: PMC6557097 DOI: 10.4103/1673-5374.255973] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Recently, we have reported that Oenanthe javanica extract (OJE) displays strong neuroprotective effect against ischemic damage after transient global cerebral ischemia. However, neuroprotective mechanisms of OJE have not been fully identified. Thus, this study investigated the neuroprotection of OJE in the hippocampal CA1 area and its anti-inflammatory activity in gerbils subjected to 5 minutes of transient global cerebral ischemia. We treated the animals by intragastrical injection of OJE (100 and 200 mg/kg) once daily for 1 week prior to transient global cerebral ischemia. Neuroprotection of OJE was observed by immunohistochemistry for neuronal nuclear antigen and histofluorescence staining for Fluoro-Jade B. Immunohistochemistry of glial fibrillary acidic protein and ionized calcium-binding adapter molecule 1 was done for astrocytosis and microgliosis, respectively. To investigate the neuroprotective mechanisms of OJE, we performed immunohistochemistry of tumor necrosis factor-alpha and interleukin-2 for pro-inflammatory function and interleukin-4 and interleukin-13 for anti-inflammatory function. When we treated the animals by intragastrical administration of 200 mg/kg of OJE, hippocampal CA1 pyramidal neurons were protected from transient global cerebral ischemia and cerebral ischemia-induced gliosis was inhibited in the ischemic hippocampal CA1 area. We also found that interleukin-4 and -13 immunoreactivities were significantly increased in pyramidal neurons of the ischemic CA1 area after OJE pretreatment, and the increased immunoreactivities were sustained in the CA1 pyramidal neurons after transient global cerebral ischemia. However, OJE pretreatment did not increase interleukin-2 and tumor necrosis factor-alpha immunoreactivities in the CA1 pyramidal neurons. Our findings suggest that pretreatment with OJE can protect neurons and attenuate gliosis from transient global cerebral ischemia via increasing expressions of interleukin-4 and -13. The experimental plan of this study was reviewed and approved by the Institutional Animal Care and Use Committee (IACUC) in Kangwon National University (approval No. KW-160802-1) on August 10, 2016.
Collapse
Affiliation(s)
- Joon Ha Park
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon, Republic of Korea
| | - In Hye Kim
- Famenity Company, Gwacheon, Geyonggi, Republic of Korea
| | - Ji Hyeon Ahn
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon, Republic of Korea
| | - Yoo Hun Noh
- Famenity Company, Gwacheon, Geyonggi, Republic of Korea
| | - Sung-Su Kim
- Famenity Company, Gwacheon, Geyonggi, Republic of Korea
| | - Tae-Kyeong Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| | - Bich-Na Shin
- Danchunok Company, Chuncheon, Gangwon, Republic of Korea
| | - Tae Heung Sim
- Danchunok Company, Chuncheon, Gangwon, Republic of Korea
| | - Hyun Sam Lee
- Danchunok Company, Chuncheon, Gangwon, Republic of Korea
| | - Jeong Hwi Cho
- Department of Histology, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeollabuk-do, Republic of Korea
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, Republic of Korea
| | - Il Jun Kang
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Gangwon, Republic of Korea
| | - Jong Dai Kim
- Division of Food Biotechnology, School of Biotechnology, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| |
Collapse
|
33
|
Comparative study of the effects of Danhong injection with different doses on ischemic stroke: A substudy of hospital-based Danhong injection registry. J TRADIT CHIN MED 2018. [DOI: 10.1016/s0254-6272(18)30992-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
34
|
Wang X, Guo J, Ning Z, Wu X. Discovery of a Natural Syk Inhibitor from Chinese Medicine through a Docking-Based Virtual Screening and Biological Assay Study. Molecules 2018; 23:molecules23123114. [PMID: 30487406 PMCID: PMC6320911 DOI: 10.3390/molecules23123114] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 11/20/2018] [Accepted: 11/26/2018] [Indexed: 12/17/2022] Open
Abstract
Spleen tyrosine kinase (Syk) is a critical target protein for treating immunoreceptor signalling-mediated allergies. In this study, a virtual screening of an in-house Chinese medicine database followed by biological assays was carried out to identify novel Syk inhibitors. A molecular docking method was employed to screen for compounds with potential Syk inhibitory activity. Then, an in vitro kinase inhibition assay was performed to verify the Syk inhibitory activity of the virtual screening hits. Subsequently, a β-hexosaminidase release assay was conducted to evaluate the anti-mast cell degranulation activity of the active compounds. Finally, tanshinone I was confirmed as a Syk inhibitor (IC50 = 1.64 μM) and exhibited anti-mast cell degranulation activity in vitro (IC50 = 2.76 μM). Docking studies showed that Pro455, Gln462, Leu377, and Lys458 were key amino acid residues for Syk inhibitory activity. This study demonstrated that tanshinone I is a Syk inhibitor with mast cell degranulation inhibitory activity. Tanshinone I may be a potential lead compound for developing effective and safe Syk-inhibiting drugs.
Collapse
Affiliation(s)
- Xing Wang
- School of Traditional Chinese Medicine, Capital Medical University, Fengtai District, Beijing 100069, China.
- Beijing Key Lab of Traditional Chinese Medicine Collateral Disease Theory Research, Capital Medical University, Fengtai District, Beijing 100069, China.
| | - Junfang Guo
- School of Traditional Chinese Medicine, Capital Medical University, Fengtai District, Beijing 100069, China.
| | - Zhongqi Ning
- School of Traditional Chinese Medicine, Capital Medical University, Fengtai District, Beijing 100069, China.
| | - Xia Wu
- School of Traditional Chinese Medicine, Capital Medical University, Fengtai District, Beijing 100069, China.
- Beijing Key Lab of Traditional Chinese Medicine Collateral Disease Theory Research, Capital Medical University, Fengtai District, Beijing 100069, China.
| |
Collapse
|
35
|
Immune Cells After Ischemic Stroke Onset: Roles, Migration, and Target Intervention. J Mol Neurosci 2018; 66:342-355. [DOI: 10.1007/s12031-018-1173-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 09/14/2018] [Indexed: 01/09/2023]
|
36
|
Park Y, Tae HJ, Cho JH, Kim IS, Ohk TG, Park CW, Moon JB, Shin MC, Lee TK, Lee JC, Park JH, Ahn JH, Kang SH, Won MH, Cho JH. The relationship between low survival and acute increase of tumor necrosis factor α expression in the lung in a rat model of asphyxial cardiac arrest. Anat Cell Biol 2018; 51:128-135. [PMID: 29984058 PMCID: PMC6026820 DOI: 10.5115/acb.2018.51.2.128] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 05/02/2018] [Accepted: 05/12/2018] [Indexed: 01/17/2023] Open
Abstract
Cardiac arrest (CA) is sudden loss of heart function and abrupt stop in effective blood flow to the body. The patients who initially achieve return of spontaneous circulation (RoSC) after CA have low survival rate. It has been known that multiorgan dysfunctions after RoSC are associated with high morbidity and mortality. Most previous studies have focused on the heart and brain in RoSC after CA. Therefore, the aim of this research was to perform serological, physiological, and histopathology study in the lung and to determine whether or how pulmonary dysfunction is associated with low survival rate after CA. Experimental animals were divided into sham-operated group (n=14 at each point in time), which was not subjected to CA operation, and CA-operated group (n=14 at each point in time), which was subjected to CA. The rats in each group were sacrificed at 6 hours, 12 hours, 24 hours, and 2 days, respectively, after RoSC. Then, pathological changes of the lungs were analyzed by hematoxylin and eosin staining, Western blot and immunohistochemistry for tumor necrosis factor α (TNF-α). The survival rate after CA was decreased with time past. We found that histopathological score and TNF-α immunoreactivity were significantly increased in the lung after CA. These results indicate that inflammation triggered by ischemia-reperfusion damage after CA leads to pulmonary injury/dysfunctions and contributes to low survival rate. In addition, the finding of increase in TNF-α via inflammation in the lung after CA would be able to utilize therapeutic or diagnostic measures in the future.
Collapse
Affiliation(s)
- Yoonsoo Park
- Department of Emergency Medicine and Institute of Medical Sciences, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Hyun-Jin Tae
- Bio Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Korea
| | - Jeong Hwi Cho
- Bio Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Korea
| | - In-Shik Kim
- Bio Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Korea
| | - Taek Geun Ohk
- Department of Emergency Medicine and Institute of Medical Sciences, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Chan Woo Park
- Department of Emergency Medicine and Institute of Medical Sciences, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Joong Bum Moon
- Department of Emergency Medicine and Institute of Medical Sciences, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Myoung Cheol Shin
- Department of Emergency Medicine and Institute of Medical Sciences, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Tae-Kyeong Lee
- Department of Neurobiology, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Jae-Chul Lee
- Department of Neurobiology, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Joon Ha Park
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, Korea
| | - Ji Hyeon Ahn
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, Korea
| | - Seok Hoon Kang
- Department of Medical Education, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Moo-Ho Won
- Department of Neurobiology, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Jun Hwi Cho
- Department of Emergency Medicine and Institute of Medical Sciences, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, Korea
| |
Collapse
|
37
|
Kim DH, Shin EA, Kim B, Shim BS, Kim SH. Reactive oxygen species-mediated phosphorylation of p38 signaling is critically involved in apoptotic effect of Tanshinone I in colon cancer cells. Phytother Res 2018; 32:1975-1982. [DOI: 10.1002/ptr.6126] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 05/11/2018] [Accepted: 05/15/2018] [Indexed: 11/10/2022]
Affiliation(s)
- Dong Hee Kim
- College of Korean Medicine; Kyung Hee University; Seoul South Korea
| | - Eun Ah Shin
- College of Korean Medicine; Kyung Hee University; Seoul South Korea
| | - Bonglee Kim
- College of Korean Medicine; Kyung Hee University; Seoul South Korea
| | - Bum Sang Shim
- College of Korean Medicine; Kyung Hee University; Seoul South Korea
| | - Sung-Hoon Kim
- College of Korean Medicine; Kyung Hee University; Seoul South Korea
| |
Collapse
|
38
|
Lee JC, Park CW, Shin MC, Cho JH, Lee HA, Kim YM, Park JH, Ahn JH, Cho JH, Tae HJ, Hwang IK, Lee TK, Won MH, Kang IJ. Tumor necrosis factor receptor 2 is required for ischemic preconditioning-mediated neuroprotection in the hippocampus following a subsequent longer transient cerebral ischemia. Neurochem Int 2018; 118:292-303. [PMID: 29777731 DOI: 10.1016/j.neuint.2018.05.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 04/26/2018] [Accepted: 05/14/2018] [Indexed: 01/08/2023]
Abstract
Tumor Necrosis Factor-α (TNF-α) is a proinflammatory cytokine implicated in neuronal damage in response to cerebral ischemia. Ischemic preconditioning (IPC) provides neuroprotection against a subsequent severer or longer transient ischemia by ischemic tolerance. Here, we focused on the role of TNF-α in IPC-mediated neuroprotection against neuronal death following a subsequent longer transient cerebral ischemia (TCI). Gerbils used in this study were randomly assigned to eight groups; sham group, TCI operated group, IPC plus (+) sham group, IPC + TCI operated group, sham + etanercept (an inhibitor of TNF-a) group, TCI + etanercept group, IPC + sham + etanercept group, and IPC + TCI + etanercept group. IPC was induced by a 2-min sublethal transient ischemia, which was operated 1 day prior to a longer (5-min) TCI. A significant death of neurons was found in the stratum pyramidale (SP) in the CA1 area (CA1) of the hippocampus 5 days after TCI; however, IPC protected SP neurons from TCI. We found that TNF-α immunoreactivity was significantly increased in CA1 pyramidal neurons in the TCI and IPC + TCI groups compared to the sham group. TNF-R1 expression in CA1 pyramidal neurons of the TCI group was also increased 1 and 2 days after TCI; however, in the IPC + TCI group, TNF-R1 expression was significantly lower than that in the TCI group. On the other hand, we did not detect TNF-R2 immunoreactivity in CA1 pyramidal neurons 1 and 2 days after TCI; meanwhile, in the IPC + TCI group, TNF-R2 expression was significantly increased compared to TNF-R2 expression at 1 and 2 days after TCI. In addition, in this group, TNF-R2 was newly expressed in pericytes, which are important cells in the blood brain barrier, from 1 day after TCI. When we treated etanercept to the IPC + TCI group, IPC-induced neuroprotection was significantly weakened. In brief, this study indicates that IPC confers neuroprotection against TCI by TNF-α signaling through TNF-R2 and suggests that the enhancement of TNF-R2 expression by IPC may be a legitimate strategy for a therapeutic intervention of TCI.
Collapse
Affiliation(s)
- Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon, 24341 Republic of Korea
| | - Chan Woo Park
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon, 24341 Republic of Korea
| | - Myoung Cheol Shin
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon, 24341 Republic of Korea
| | - Jun Hwi Cho
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon, 24341 Republic of Korea
| | - Hyang-Ah Lee
- Department of Obstetrics and Gynecology, School of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Joon Ha Park
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon, 24252 Republic of Korea
| | - Ji Hyeon Ahn
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon, 24252 Republic of Korea
| | - Jeong Hwi Cho
- Bio-Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Chonbuk, 54596 Republic of Korea
| | - Hyun-Jin Tae
- Bio-Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Chonbuk, 54596 Republic of Korea
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826 Republic of Korea
| | - Tae-Kyeong Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon, 24341 Republic of Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon, 24341 Republic of Korea.
| | - Il Jun Kang
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Gangwon, 24252 Republic of Korea.
| |
Collapse
|
39
|
Zhu Y, Tang Q, Wang G, Han R. Tanshinone IIA Protects Hippocampal Neuronal Cells from Reactive Oxygen Species Through Changes in Autophagy and Activation of Phosphatidylinositol 3-Kinase, Protein Kinas B, and Mechanistic Target of Rapamycin Pathways. Curr Neurovasc Res 2018; 14:132-140. [PMID: 28260507 PMCID: PMC5543574 DOI: 10.2174/1567202614666170306105315] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 02/18/2017] [Accepted: 02/20/2017] [Indexed: 12/29/2022]
Abstract
Background: Tanshinone IIA is a key active ingredient of danshen, which is derived from the dried root or rhizome of Salviae miltiorrhizae Bge. The tanshinone IIA has protective effects against the focal cerebral ischemic injury. However, the underlying mechanisms remain unclear. Methods: An in vitro model of cerebral ischemia was established by subjecting cultures of hippocampal neuronal cells to oxygen-glucose deprivation followed by reperfusion (OGD/R). The probes of 5-(and-6)-chloromethyl-2’,7’-dichlorodihydrofluorescein diacetate, acetyl ester (CM-H2DCFDA) and 5’,6,6’-tetrachloro-1,1’,3,3’-tetraethylbenzimidazolylcarbocyanine,iodide (JC-1) were used to determine the mitochondrial membrane potential (MMP) and reactive oxygen species (ROS) production. Western-blot was used to detect the expression of proteins in HT-22 cells. Results: The results of cell proliferative assays showed that the tanshinone IIA attenuated OGD/R-mediated neuronal cell death, with the evidence of increased cell viability. In addition, OGD/R exposure led to increase the levels of intracellular reactive oxygen species (ROS), which were significantly suppressed by tanshinone IIA treatment. Furthermore, tanshinone IIA treatment inhibited elevations in MMP and autophagy following exposure to OGD/R. Additionally, OGD/R promoted cell death with concomitant inhibiting phosphatidylinositol-3-kinase (PI3K)/protein kinase B (Akt)/ mammalian target of Rapamycin (mTOR) pathway, which was reversed by tanshinone IIA. Conclusion: These results suggest that the tanshinone IIA protects against OGD/R-mediated cell death in HT-22 cells, in part, due to activating PI3K/Akt/mTOR pathway.
Collapse
Affiliation(s)
- Yingchun Zhu
- Department of Neurology Disease, the Affiliated Anhui Provincial Hospital of Anhui Medical University, Hefei 230022, Anhui, China
| | - Qiqiang Tang
- Department of Neurology Disease, the Affiliated Anhui Provincial Hospital of Anhui Medical University, Hefei 230022, Anhui, China
| | - Guopin Wang
- Department of Neurology Disease, the Affiliated Anhui Provincial Hospital of Anhui Medical University, Hefei 230022, Anhui, China
| | - Ruodong Han
- Department of Intensive Care Division, The People's Hospital of Bozhou, Bozhou 236800, Anhui, China
| |
Collapse
|
40
|
Han JY, Li Q, Ma ZZ, Fan JY. Effects and mechanisms of compound Chinese medicine and major ingredients on microcirculatory dysfunction and organ injury induced by ischemia/reperfusion. Pharmacol Ther 2017; 177:146-173. [PMID: 28322971 DOI: 10.1016/j.pharmthera.2017.03.005] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Microcirculation dysfunction and organ injury after ischemia and reperfusion (I/R) result from a complex pathologic process consisting of multiple links, with metabolism impairment in the ischemia phase and oxidative stress in the reperfusion phase as initiators, and any treatment targeting a single link is insufficient to cope with this. Compound Chinese medicine (CCM) has been applied in clinics in China and some Asian nations for >2000years. Studies over the past decades revealed the protective and therapeutic effect of CCMs and major ingredients on I/R-induced microcirculatory dysfunction and tissue injury in the heart, brain, liver, intestine, and so on. CCM contains diverse bioactive components with potential for energy metabolism regulation; antioxidant effect; inhibiting inflammatory cytokines release; adhesion molecule expression in leukocyte, platelet, and vascular endothelial cells; and the protection of thrombosis, albumin leakage, and mast cell degranulation. This review covers the major works with respect to the effects and underlying mechanisms of CCM and its ingredients on microcirculatory dysfunction and organ injury after I/R, providing novel ideas for dealing with this threat.
Collapse
Affiliation(s)
- Jing-Yan Han
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing 100191, China; Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing 100191, China; Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing 100191, China; Beijing Microvascular Institute of Integration of Chinese and Western Medicine, Beijing, China.
| | - Quan Li
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing 100191, China; Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing 100191, China; Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing 100191, China; Beijing Microvascular Institute of Integration of Chinese and Western Medicine, Beijing, China
| | - Zhi-Zhong Ma
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Jing-Yu Fan
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing 100191, China; Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing 100191, China; Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing 100191, China; Beijing Microvascular Institute of Integration of Chinese and Western Medicine, Beijing, China
| |
Collapse
|
41
|
Tae HJ, Kang IJ, Lee TK, Cho JH, Lee JC, Shin MC, Kim YS, Cho JH, Kim JD, Ahn JH, Park JH, Kim IS, Lee HA, Kim YH, Won MH, Lee YJ. Neuronal injury and tumor necrosis factor-alpha immunoreactivity in the rat hippocampus in the early period of asphyxia-induced cardiac arrest under normothermia. Neural Regen Res 2017; 12:2007-2013. [PMID: 29323039 PMCID: PMC5784348 DOI: 10.4103/1673-5374.221157] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Low survival rate occurs in patients who initially experience a spontaneous return of circulation after cardiac arrest (CA). In this study, we induced asphyxial CA in adult male Sprague-Daley rats, maintained their body temperature at 37 ± 0.5°C, and then observed the survival rate during the post-resuscitation phase. We examined neuronal damage in the hippocampus using cresyl violet (CV) and Fluore-Jade B (F-J B) staining, and pro-inflammatory response using ionized calcium-binding adapter molecule 1 (Iba-1), glial fibrillary acidic protein (GFAP), and tumor necrosis factor-alpha (TNF-α) immunohistochemistry in the hippocampus after asphyxial CA in rats under normothermia. Our results show that the survival rate decreased gradually post-CA (about 63% at 6 hours, 37% at 1 day, and 8% at 2 days post-CA). Rats were sacrificed at these points in time post-CA, and no neuronal damage was found in the hippocampus until 1 day post-CA. However, some neurons in the stratum pyramidale of the CA region in the hippocampus were dead 2 days post-CA. Iba-1 immunoreactive microglia in the CA1 region did not change until 1 day post-CA, and they were activated (enlarged cell bodies with short and thicken processes) in all layers 2 days post-CA. Meanwhile, GFAP-immunoreactive astrocytes did not change significantly until 2 days post-CA. TNF-α immunoreactivity decreased significantly in neurons of the stratum pyramidale in the CA1 region 6 hours post-CA, decreased gradually until 1 day post-CA, and increased significantly again 2 days post-CA. These findings suggest that low survival rate of normothermic rats in the early period of asphyxia-induced CA is related to increased TNF-α immunoreactivity, but not to neuronal damage in the hippocampal CA1 region.
Collapse
Affiliation(s)
- Hyun-Jin Tae
- Bio-Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, South Korea
| | - Il Jun Kang
- Department of Food Science and Nutrition, Hallym University, Chuncheon, South Korea
| | - Tae-Kyeong Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Jeong Hwi Cho
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Myoung Cheol Shin
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Yoon Sung Kim
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon; Department of Emergency Medicine, Samcheok Medical Center, Samcheok, South Korea
| | - Jun Hwi Cho
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Jong-Dai Kim
- Division of Food Biotechnology, School of Biotechnology, Kangwon National University, Chuncheon, South Korea
| | - Ji Hyeon Ahn
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, South Korea
| | - Joon Ha Park
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, South Korea
| | - In-Shik Kim
- Bio-Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, South Korea
| | - Hyang-Ah Lee
- Department of Obstetrics and Gynecology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Yang Hee Kim
- Department of Surgery, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Young Joo Lee
- Department of Emergency Medicine, Seoul Hospital, College of Medicine, Sooncheonhyang University, Seoul, South Korea
| |
Collapse
|
42
|
Tanshinones and mental diseases: from chemistry to medicine. Rev Neurosci 2016; 27:777-791. [DOI: 10.1515/revneuro-2016-0012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 06/03/2016] [Indexed: 11/15/2022]
Abstract
AbstractThe prevalence of mental diseases, especially neurodegenerative disorders, is ever-increasing, while treatment options for such disorders are limited and insufficient. In this scarcity of available medication, it is a feasible strategy to search for potential drugs among natural compounds, such as those found in plants. One such plant source is the root of Chinese sage, Salvia miltiorrhiza Bunge (Labiatae), which contains several compounds reported to possess neuroprotective activities. The most important of these compounds are tanshinones, which have been reported to possess ameliorative activity against a myriad of mental diseases such as Alzheimer’s disease, cerebral ischemia/reperfusion injury, and glioma, along with promoting neuronal differentiation and manifesting antinociceptive and anticonvulsant outcomes. This review offers a critical evaluation of the utility of tanshinones to treat mental illnesses, and sheds light on the underlying mechanisms through which these naturally occurring compounds confer neuroprotection.
Collapse
|
43
|
Tanshinone I Attenuates the Effects of a Challenge with H 2O 2 on the Functions of Tricarboxylic Acid Cycle and Respiratory Chain in SH-SY5Y Cells. Mol Neurobiol 2016; 54:7858-7868. [PMID: 27848206 DOI: 10.1007/s12035-016-0267-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 10/30/2016] [Indexed: 12/24/2022]
Abstract
Tanshinone I (T-I; C18H12O3) is a cytoprotective molecule. T-I has been viewed as an antioxidant and anti-inflammatory agent exerting neuroprotective actions in several experimental models. Nonetheless, the mechanisms underlying the beneficial effects of T-I in mammalian cells are not completely understood yet. Mitochondrial dysfunction has been associated with several neurodegenerative diseases which remain uncured. Therefore, there is increasing interest in compounds that may be used in the prevention or treatment of those pathologies. Since T-I presents an antioxidant capacity, we investigated here whether and how this compound would prevent mitochondrial impairment in SH-SY5Y cells exposed to hydrogen peroxide (H2O2), which has been involved in the triggering of deleterious effects in several experimental models mimicking neurodegenerative processes. We found that a pretreatment with T-I at 2.5 μM for 2 h suppressed the pro-oxidant effects of H2O2 on mitochondrial membranes. Furthermore, T-I prevented the H2O2-elicited inhibition of the tricarboxylic acid (TCA) cycle enzymes (aconitase, α-ketoglutarate dehydrogenase, and succinate dehydrogenase) and of the mitochondrial complexes I and V. T-I also abrogated the mitochondrial depolarization and the mitochondrial failure to produce ATP in cells exposed to H2O2. T-I upregulated the levels of reduced glutathione (GSH) in the mitochondria of SH-SY5Y cells. T-I induced mitochondrial protection, at least in part, by activating the nuclear factor erythroid 2-related factor 2 (Nrf2), because silencing of Nrf2 by using small interference RNA (SiRNA) blocked these effects. Therefore, T-I afforded mitochondrial protection (involving both redox and bioenergetics-related aspects) against H2O2 through the activation of Nrf2.
Collapse
|
44
|
Park JH, Shin BN, Ahn JH, Cho JH, Kim IH, Kim DW, Won MH, Hong S, Cho JH, Lee CH. Ischemia-Induced Changes of PRAS40 and p-PRAS40 Immunoreactivities in the Gerbil Hippocampal CA1 Region After Transient Cerebral Ischemia. Cell Mol Neurobiol 2016; 36:821-8. [PMID: 26526334 PMCID: PMC11482446 DOI: 10.1007/s10571-015-0265-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 08/28/2015] [Indexed: 12/24/2022]
Abstract
Proline-rich Akt substrate of 40-kDa (PRAS40) is one of the important interactive linkers between Akt and mTOR signaling pathways. The increase of PRAS40 is related with the reduction of brain damage induced by cerebral ischemia. In the present study, we investigated time-dependent changes in PRAS40 and phospho-PRAS40 (p-PRAS40) immunoreactivities in the hippocampal CA1 region of the gerbil after 5 min of transient cerebral ischemia. PRAS40 immunoreactivity in the CA1 region was decreased in pyramidal neurons from 12 h after ischemic insult in a time-dependent manner, and, at 5 days post-ischemia, PRAS40 immunoreactivity was newly expressed in astrocytes. p-PRAS40 immunoreactivity in the CA1 pyramidal neurons was hardly found 12 h and apparently detected again 1 and 2 days after ischemic insult. At 5 days post-ischemia, p-PRAS40 immunoreactivity in the CA1 pyramidal neurons was not found. These results indicate that ischemia-induced changes in PRAS40 and p-PRAS40 immunoreactivities in CA1 pyramidal neurons and astrocytes may be closely associated with delayed neuronal death in the hippocampal CA1 region following transient cerebral ischemia.
Collapse
Affiliation(s)
- Joon Ha Park
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 200-701, South Korea
| | - Bich Na Shin
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, 200-702, South Korea
| | - Ji Hyeon Ahn
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 200-701, South Korea
| | - Jeong-Hwi Cho
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 200-701, South Korea
| | - In Hye Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 200-701, South Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, College of Dentistry, and Research Institute of Oral Sciences, Kangnung-Wonju National University, Gangneung, 210-702, South Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 200-701, South Korea
| | - Seongkweon Hong
- Department of Surgery, School of Medicine, Kangwon National University, Chuncheon, 200-701, South Korea
| | - Jun Hwi Cho
- Department of Emergency Medicine, College of Medicine, School of Medicine, Kangwon National University, Chuncheon, 200-701, South Korea.
| | - Choong-Hyun Lee
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan, 330-714, South Korea.
| |
Collapse
|
45
|
Fu B, Guo S, Yu X, Chang H, Wang W. Astragalus Salvia Granules to Benefit the Qi (Qishen Yiqi Keli) protects H9C2 cardiomyocytes by suppressing oxidative stress. JOURNAL OF TRADITIONAL CHINESE MEDICAL SCIENCES 2016. [DOI: 10.1016/j.jtcms.2016.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
46
|
Chang CC, Lee YC, Lin CC, Chang CH, Chiu CD, Chou LW, Sun MF, Yen HR. Characteristics of traditional Chinese medicine usage in patients with stroke in Taiwan: A nationwide population-based study. JOURNAL OF ETHNOPHARMACOLOGY 2016; 186:311-321. [PMID: 27090345 DOI: 10.1016/j.jep.2016.04.018] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 04/11/2016] [Accepted: 04/12/2016] [Indexed: 06/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Stroke has been the leading causes of death worldwide. Traditional Chinese medicine (TCM) has been used for stoke patients for thousands of years. This study aimed to investigate TCM usage and prescription patterns in stroke patients in Taiwan. MATERIALS AND METHODS We analyzed a random sample of one million individuals representing the 23 million enrollees selected from the National Health Insurance Research Database in Taiwan. Demographic characteristics, TCM usage, prescription patterns and mortality rate among stroke patients were analyzed. RESULTS We identified 23,816 patients who were newly diagnosed with stroke between 2001 and 2009 by their diagnostic codes (ICD-9-CM 430-438). Among them, 4302 patients had hemorrhagic stroke while 19,514 patients had ischemic stroke. Overall, 12% of the stroke patients (n=2862) were TCM users. The median interval between stroke onset to the first TCM consultation is 12.2 months. Among the TCM users, more than half (52.7%) of the patients received both Chinese herbal remedies and acupuncture/traumatology treatment. Bu-yang-huan-wu-tang and Dan-shen (Radix Salviae Miltiorrhizae; Salvia miltiorrhiza Bunge) was the most commonly prescribed Chinese herbal formula and single herb, respectively. TCM users had a higher incidence rate ratio in myalgia, myositis, fasciitis and insomnia than non-TCM users. Mental disorders such as anxiety and depression are common in both TCM and non-TCM users. Comparing with the non-TCM users, the TCM users had a lower mortality rate (adjusted hazard ratios were 0.44 in overall stroke, 0.50 in ischemic stroke and 0.25 in hemorrhagic stroke). CONCLUSION Adjunctive TCM use may reduce the risk of mortality rate among stroke patients. Bu-yang-huan-wu-tang and Dan-shen are the most common prescribed Chinese herbal formula and single herb for stroke patients, respectively. Future study investigating the anti-inflammatory and neuroprotective efficacy of Bu-yang-huan-wu-tang and Dan-shen in stroke is warranted.
Collapse
Affiliation(s)
- Chia-Chi Chang
- Department of Chinese Medicine, China Medical University Hospital, Taichung 404, Taiwan; Research Center for Traditional Chinese Medicine, Department of Medical Research, China Medical University Hospital, Taichung 404, Taiwan.
| | - Yu-Chen Lee
- Department of Chinese Medicine, China Medical University Hospital, Taichung 404, Taiwan; Research Center for Chinese Medicine & Acupuncture, China Medical University, Taichung 404, Taiwan; Graduate Institute of Acupuncture Science, China Medical University, Taichung 404, Taiwan.
| | - Che-Chen Lin
- Health Data Management Office, China Medical University Hospital, Taichung 404, Taiwan.
| | - Chin-Hsien Chang
- Department of Traditional Chinese Medicine, En Chu Kong Hospital, New Taipei City 237, Taiwan; Department of Cosmetic Science, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan.
| | - Cheng-Di Chiu
- Department of Neurosurgery, China Medical University Hospital, Taichung 404, Taiwan; School of Medicine, China Medical University, Taichung 404, Taiwan.
| | - Li-Wei Chou
- Research Center for Chinese Medicine & Acupuncture, China Medical University, Taichung 404, Taiwan; Graduate Institute of Acupuncture Science, China Medical University, Taichung 404, Taiwan; School of Chinese Medicine, China Medical University, Taichung 404, Taiwan; Department of Physical Medicine and Rehabilitation, China Medical University Hospital, Taichung 404, Taiwan.
| | - Mao-Feng Sun
- Department of Chinese Medicine, China Medical University Hospital, Taichung 404, Taiwan; Research Center for Chinese Medicine & Acupuncture, China Medical University, Taichung 404, Taiwan; School of Chinese Medicine, China Medical University, Taichung 404, Taiwan.
| | - Hung-Rong Yen
- Department of Chinese Medicine, China Medical University Hospital, Taichung 404, Taiwan; Research Center for Traditional Chinese Medicine, Department of Medical Research, China Medical University Hospital, Taichung 404, Taiwan; Research Center for Chinese Medicine & Acupuncture, China Medical University, Taichung 404, Taiwan; School of Chinese Medicine, China Medical University, Taichung 404, Taiwan.
| |
Collapse
|
47
|
Park JH, Cho JH, Kim IH, Ahn JH, Lee JC, Chen BH, Shin BN, Tae HJ, Yoo KY, Hong S, Kang IJ, Won MH, Kim JD. Oenanthe Javanica Extract Protects Against Experimentally Induced Ischemic Neuronal Damage via its Antioxidant Effects. Chin Med J (Engl) 2016; 128:2932-7. [PMID: 26521793 PMCID: PMC4756874 DOI: 10.4103/0366-6999.168063] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Water dropwort (Oenanthe javanica) as a popular traditional medicine in Asia shows various biological properties including antioxidant activity. In this study, we firstly examined the neuroprotective effect of Oenanthe javanica extract (OJE) in the hippocampal cornus ammonis 1 region (CA1 region) of the gerbil subjected to transient cerebral ischemia. METHODS Gerbils were established by the occlusion of common carotid arteries for 5 min. The neuroprotective effect of OJE was estimated by cresyl violet staining. In addition, 4 antioxidants (copper, zinc superoxide dismutase [SOD], manganese SOD, catalase, and glutathione peroxidase) immunoreactivities were investigated by immunohistochemistry. RESULTS Pyramidal neurons in the CA1 region showed neuronal death at 5 days postischemia; at this point in time, all antioxidants immunoreactivities disappeared in CA1 pyramidal neurons and showed in many nonpyramidal cells. Treatment with 200 mg/kg, not 100 mg/kg, OJE protected CA1 pyramidal neurons from ischemic damage. In addition, 200 mg/kg OJE treatment increased or maintained antioxidants immunoreactivities. Especially, among the antioxidants, glutathione peroxidase immunoreactivity was effectively increased in the CA1 pyramidal neurons of the OJE-treated sham-operated and ischemia-operated groups. CONCLUSION Our present results indicate that treatment with OJE can protect neurons from transient ischemic damage and that the neuroprotective effect may be closely associated with increased or maintained intracellular antioxidant enzymes by OJE.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 200-701, South Korea
| | - Jong-Dai Kim
- Division of Food Biotechnology, School of Biotechnology, Kangwon National University, Chuncheon 200-701, South Korea
| |
Collapse
|
48
|
Choi HY, Park JH, Chen BH, Shin BN, Lee YL, Kim IH, Cho JH, Lee TK, Lee JC, Won MH, Ahn JH, Tae HJ, Yan BC, Hwang IK, Cho JH, Kim YM, Kim SK. Increases of Catalase and Glutathione Peroxidase Expressions by Lacosamide Pretreatment Contributes to Neuroprotection Against Experimentally Induced Transient Cerebral Ischemia. Neurochem Res 2016; 41:2380-90. [DOI: 10.1007/s11064-016-1951-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 05/02/2016] [Accepted: 05/07/2016] [Indexed: 12/29/2022]
|
49
|
Yoo KY, Kim IH, Cho JH, Ahn JH, Park JH, Lee JC, Tae HJ, Kim DW, Kim JD, Hong S, Won MH, Kang IJ. Neuroprotection of Chrysanthemum indicum Linne against cerebral ischemia/reperfusion injury by anti-inflammatory effect in gerbils. Neural Regen Res 2016; 11:270-7. [PMID: 27073380 PMCID: PMC4810991 DOI: 10.4103/1673-5374.177735] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
In this study, we tried to verify the neuroprotective effect of Chrysanthemum indicum Linne (CIL) extract, which has been used as a botanical drug in East Asia, against ischemic damage and to explore the underlying mechanism involving the anti-inflammatory approach. A gerbil was given CIL extract for 7 consecutive days followed by bilateral carotid artery occlusion to make a cerebral ischemia/reperfusion model. Then, we found that CIL extracts protected pyramidal neurons in the hippocampal CA1 region (CA1) from ischemic damage using neuronal nucleus immunohistochemistry and Fluoro-Jade B histofluorescence. Accordingly, interleukin-13 immunoreactivities in the CA1 pyramidal neurons of CIL-pretreated animals were maintained or increased after cerebral ischemia/reperfusion. These findings indicate that the pre-treatment of CIL can attenuate neuronal damage/death in the brain after cerebral ischemia/reperfusion via an anti-inflammatory approach.
Collapse
Affiliation(s)
- Ki-Yeon Yoo
- Department of Oral Anatomy, College of Dentistry and Research Institute of Oral Science, Gangneung-Wonju National University, Gangneung, Republic of Korea
| | - In Hye Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Jeong-Hwi Cho
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Ji Hyeon Ahn
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Joon Ha Park
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Hyun-Jin Tae
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Republic of Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, College of Dentistry and Research Institute of Oral Science, Gangneung-Wonju National University, Gangneung, Republic of Korea
| | - Jong-Dai Kim
- Division of Food Biotechnology, School of Biotechnology, Kangwon National University, Chuncheon, Republic of Korea
| | - Seongkweon Hong
- Department of Surgery, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Il Jun Kang
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Republic of Korea
| |
Collapse
|
50
|
Chen BH, Park JH, Cho JH, Kim IH, Lee JC, Lee TK, Ahn JH, Tae HJ, Shin BN, Kim JD, Kang IJ, Won MH, Lee YL. Tanshinone I Enhances Neurogenesis in the Mouse Hippocampal Dentate Gyrus via Increasing Wnt-3, Phosphorylated Glycogen Synthase Kinase-3β and β-Catenin Immunoreactivities. Neurochem Res 2016; 41:1958-68. [PMID: 27053301 DOI: 10.1007/s11064-016-1906-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 03/26/2016] [Accepted: 03/30/2016] [Indexed: 12/17/2022]
Abstract
Tanshinone I (TsI), a lipophilic diterpene extracted from Danshan (Radix Salvia miltiorrhizae), exerts neuroprotection in cerebrovascular diseases including transient ischemic attack. In this study, we examined effects of TsI on cell proliferation and neuronal differentiation in the subgranular zone (SGZ) of the mouse dentate gyrus (DG) using Ki-67, BrdU and doublecortin (DCX) immunohistochemistry. Mice were treated with 1 and 2 mg/kg TsI for 28 days. In the 1 mg/kg TsI-treated-group, distribution patterns of BrdU, Ki-67 and DCX positive ((+)) cells in the SGZ were similar to those in the vehicle-treated-group. However, in the 2 mg/kg TsI-treated-group, double labeled BrdU(+)/NeuN(+) cells, which are mature neurons, as well as Ki-67(+), DCX(+) and BrdU(+) cells were significantly increased compared with those in the vehicle-treated-group. On the other hand, immunoreactivities and protein levels of Wnt-3, β-catenin and serine-9-glycogen synthase kinase-3β (p-GSK-3β), which are related with morphogenesis, were significantly increased in the granule cell layer of the DG only in the 2 mg/kg TsI-treated-group. Therefore, these findings indicate that TsI can promote neurogenesis in the mouse DG and that the neurogenesis is related with increases of Wnt-3, p-GSK-3β and β-catenin immunoreactivities.
Collapse
Affiliation(s)
- Bai Hui Chen
- Department of Physiology, Institute of Neurodegeneration and Neuroregeneration, College of Medicine, Hallym University, Chuncheon, 24252, South Korea
| | - Joon Ha Park
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 24341, South Korea
| | - Jeong Hwi Cho
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 24341, South Korea
| | - In Hye Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 24341, South Korea
| | - Jae Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 24341, South Korea
| | - Tae-Kyeong Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 24341, South Korea
| | - Ji Hyeon Ahn
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Hyun Jin Tae
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Bich Na Shin
- Department of Physiology, Institute of Neurodegeneration and Neuroregeneration, College of Medicine, Hallym University, Chuncheon, 24252, South Korea
| | - Jong-Dai Kim
- Division of Food Biotechnology, School of Biotechnology, Kangwon National University, Chuncheon, 24341, South Korea
| | - Il Jun Kang
- Department of Food Science and Nutrition, Hallym University, Chuncheon, 24252, South Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 24341, South Korea.
| | - Yun Lyul Lee
- Department of Physiology, Institute of Neurodegeneration and Neuroregeneration, College of Medicine, Hallym University, Chuncheon, 24252, South Korea.
| |
Collapse
|